1
|
Zhu X, Gao C, Peng B, Xue J, Xia D, Yang L, Zhang J, Gao X, Hu Y, Lin S, Gong P, Xu X. FOXP1 phosphorylation antagonizes its O-GlcNAcylation in regulating ATR activation in response to replication stress. EMBO J 2025; 44:457-483. [PMID: 39623140 DOI: 10.1038/s44318-024-00323-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 11/13/2024] [Accepted: 11/18/2024] [Indexed: 01/15/2025] Open
Abstract
ATR signaling is essential in sensing and responding to the replication stress; as such, any defects can impair cellular function and survival. ATR itself is activated via tightly regulated mechanisms. Here, we identify FOXP1, a forkhead-box-containing transcription factor, as a regulator coordinating ATR activation. We show that, unlike its role as a transcription factor, FOXP1 functions as a scaffold and directly binds to RPA-ssDNA and ATR-ATRIP complexes, facilitating the recruitment and activation of ATR. This process is regulated by FOXP1 O-GlcNAcylation, which represses its interaction with ATR, while CHK1-mediated phosphorylation of FOXP1 inhibits its O-GlcNAcylation upon replication stress. Supporting the physiological relevance of this loop, we find pathogenic FOXP1 mutants identified in various tumor tissues with compromised ATR activation and stalled replication fork stability. We thus conclude that FOXP1 may serve as a potential chemotherapeutic target in related tumors.
Collapse
Affiliation(s)
- Xuefei Zhu
- Carson International Cancer Center & Department of General Surgery & Institute of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Shenzhen University General Hospital, Shenzhen University Medical School, 518060, Shenzhen, Guangdong, China.
- Guangdong Key Laboratory for Genome Stability & Disease Prevention and Marshall Laboratory of Biomedical Engineering, Shenzhen University Medical School, 518060, Shenzhen, Guangdong, China.
| | - Congwen Gao
- Guangdong Key Laboratory for Genome Stability & Disease Prevention and Marshall Laboratory of Biomedical Engineering, Shenzhen University Medical School, 518060, Shenzhen, Guangdong, China
- College of Life Sciences, Institute of Life Sciences and Green Development, Hebei University, 071002, Baoding, China
| | - Bin Peng
- Carson International Cancer Center & Department of General Surgery & Institute of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Shenzhen University General Hospital, Shenzhen University Medical School, 518060, Shenzhen, Guangdong, China
- Guangdong Key Laboratory for Genome Stability & Disease Prevention and Marshall Laboratory of Biomedical Engineering, Shenzhen University Medical School, 518060, Shenzhen, Guangdong, China
| | - Jingwei Xue
- Guangdong Key Laboratory for Genome Stability & Disease Prevention and Marshall Laboratory of Biomedical Engineering, Shenzhen University Medical School, 518060, Shenzhen, Guangdong, China
| | - Donghui Xia
- Guangdong Key Laboratory for Genome Stability & Disease Prevention and Marshall Laboratory of Biomedical Engineering, Shenzhen University Medical School, 518060, Shenzhen, Guangdong, China
- Shenzhen University General Hospital-Dehua Hospital Joint Research Center on Precision Medicine (sgh-dhhCPM), Dehua Hospital, Dehua, 362500, Quanzhou, China
- State Key Laboratory of Agro-biotechnology and MOA Key Laboratory of Soil Microbiology, College of Biological Sciences, China Agricultural University, 100193, Beijing, China
| | - Liu Yang
- Guangdong Key Laboratory for Genome Stability & Disease Prevention and Marshall Laboratory of Biomedical Engineering, Shenzhen University Medical School, 518060, Shenzhen, Guangdong, China
| | - Jiexiang Zhang
- Guangdong Key Laboratory for Genome Stability & Disease Prevention and Marshall Laboratory of Biomedical Engineering, Shenzhen University Medical School, 518060, Shenzhen, Guangdong, China
- Shenzhen University-Friedrich Schiller Universität Jena Joint PhD Program in Biomedical Sciences, Shenzhen University School of Medicine, 518060, Shenzhen, Guangdong, China
| | - Xinrui Gao
- Guangdong Key Laboratory for Genome Stability & Disease Prevention and Marshall Laboratory of Biomedical Engineering, Shenzhen University Medical School, 518060, Shenzhen, Guangdong, China
| | - Yilin Hu
- Life Sciences Institute, Zhejiang University, 310058, Hangzhou, China
| | - Shixian Lin
- Life Sciences Institute, Zhejiang University, 310058, Hangzhou, China
| | - Peng Gong
- Carson International Cancer Center & Department of General Surgery & Institute of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Shenzhen University General Hospital, Shenzhen University Medical School, 518060, Shenzhen, Guangdong, China.
| | - Xingzhi Xu
- Carson International Cancer Center & Department of General Surgery & Institute of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Shenzhen University General Hospital, Shenzhen University Medical School, 518060, Shenzhen, Guangdong, China.
- Guangdong Key Laboratory for Genome Stability & Disease Prevention and Marshall Laboratory of Biomedical Engineering, Shenzhen University Medical School, 518060, Shenzhen, Guangdong, China.
| |
Collapse
|
2
|
Edirisinghe O, Ternier G, Alraawi Z, Suresh Kumar TK. Decoding FGF/FGFR Signaling: Insights into Biological Functions and Disease Relevance. Biomolecules 2024; 14:1622. [PMID: 39766329 DOI: 10.3390/biom14121622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2024] [Revised: 12/06/2024] [Accepted: 12/13/2024] [Indexed: 01/11/2025] Open
Abstract
Fibroblast Growth Factors (FGFs) and their cognate receptors, FGFRs, play pivotal roles in a plethora of biological processes, including cell proliferation, differentiation, tissue repair, and metabolic homeostasis. This review provides a comprehensive overview of FGF-FGFR signaling pathways while highlighting their complex regulatory mechanisms and interconnections with other signaling networks. Further, we briefly discuss the FGFs involvement in developmental, metabolic, and housekeeping functions. By complementing current knowledge and emerging research, this review aims to enhance the understanding of FGF-FGFR-mediated signaling and its implications for health and disease, which will be crucial for therapeutic development against FGF-related pathological conditions.
Collapse
Affiliation(s)
- Oshadi Edirisinghe
- Cell and Molecular Biology Program, University of Arkansas, Fayetteville, AR 72701, USA
| | - Gaëtane Ternier
- Department of Chemistry and Biochemistry, University of Arkansas, Fayetteville, AR 72701, USA
| | - Zeina Alraawi
- Department of Chemistry and Biochemistry, University of Arkansas, Fayetteville, AR 72701, USA
| | - Thallapuranam Krishnaswamy Suresh Kumar
- Cell and Molecular Biology Program, University of Arkansas, Fayetteville, AR 72701, USA
- Department of Chemistry and Biochemistry, University of Arkansas, Fayetteville, AR 72701, USA
| |
Collapse
|
3
|
Hasan MR, Kump AJ, Stepaniak EC, Panta M, Shashidhar K, Katariya R, Sabbir MK, Schwab KR, Inlow MH, Chen Y, Ahmad SM. Genome-Wide Expression Profiling and Phenotypic Analysis of Downstream Targets Identify the Fox Transcription Factor Jumeau as a Master Regulator of Cardiac Progenitor Cell Division. Int J Mol Sci 2024; 25:12933. [PMID: 39684645 DOI: 10.3390/ijms252312933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 11/24/2024] [Accepted: 11/26/2024] [Indexed: 12/18/2024] Open
Abstract
Forkhead box (Fox) transcription factors (TFs) mediate multiple conserved cardiogenic processes in both mammals and Drosophila. Our prior work identified the roles of two Drosophila Fox genes, jumeau (jumu) and Checkpoint suppressor 1-like (CHES-1-like), in cardiac progenitor cell specification and division, and in the proper positioning of cardiac cell subtypes. Fox TF binding sites are also significantly enriched in the enhancers of genes expressed in the heart, suggesting that these genes may play a core regulatory role in one or more of these cardiogenic processes. We identified downstream targets of Jumu by comparing transcriptional expression profiles of flow cytometry-sorted mesodermal cells from wild-type embryos and embryos completely lacking the jumu gene and found that genes with functional annotation and ontological features suggesting roles in cell division were overrepresented among Jumu targets. Phenotypic analysis of a subset of these targets identified 21 jumu-regulated genes that mediate cardiac progenitor cell division, one of which, Retinal Homeobox (Rx), was characterized in more detail. Finally, the observation that many of these 21 genes and/or their orthologs exhibit genetic or physical interactions among themselves indicates that Jumu is a master regulator acting as a hub of a cardiac progenitor cell division-mediating network.
Collapse
Affiliation(s)
- M Rezaul Hasan
- Department of Biology, Indiana State University, Terre Haute, IN 47809, USA
- The Center for Genomic Advocacy, Indiana State University, Terre Haute, IN 47809, USA
- Rich and Robin Porter Cancer Research Center, Indiana State University, Terre Haute, IN 47809, USA
| | - Andrew J Kump
- Department of Biology, Indiana State University, Terre Haute, IN 47809, USA
- The Center for Genomic Advocacy, Indiana State University, Terre Haute, IN 47809, USA
- Rich and Robin Porter Cancer Research Center, Indiana State University, Terre Haute, IN 47809, USA
| | - Evelyn C Stepaniak
- Department of Biology, Indiana State University, Terre Haute, IN 47809, USA
- The Center for Genomic Advocacy, Indiana State University, Terre Haute, IN 47809, USA
- Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Manoj Panta
- Department of Biology, Indiana State University, Terre Haute, IN 47809, USA
- The Center for Genomic Advocacy, Indiana State University, Terre Haute, IN 47809, USA
| | - Kuncha Shashidhar
- Department of Biology, Indiana State University, Terre Haute, IN 47809, USA
- The Center for Genomic Advocacy, Indiana State University, Terre Haute, IN 47809, USA
- Rich and Robin Porter Cancer Research Center, Indiana State University, Terre Haute, IN 47809, USA
| | - Rajnandani Katariya
- Department of Biology, Indiana State University, Terre Haute, IN 47809, USA
- The Center for Genomic Advocacy, Indiana State University, Terre Haute, IN 47809, USA
- Rich and Robin Porter Cancer Research Center, Indiana State University, Terre Haute, IN 47809, USA
| | - Mofazzal K Sabbir
- Department of Biology, Indiana State University, Terre Haute, IN 47809, USA
- The Center for Genomic Advocacy, Indiana State University, Terre Haute, IN 47809, USA
| | - Kristopher R Schwab
- Department of Biology, Indiana State University, Terre Haute, IN 47809, USA
- The Center for Genomic Advocacy, Indiana State University, Terre Haute, IN 47809, USA
- Rich and Robin Porter Cancer Research Center, Indiana State University, Terre Haute, IN 47809, USA
| | - Mark H Inlow
- The Center for Genomic Advocacy, Indiana State University, Terre Haute, IN 47809, USA
- Department of Mathematical Sciences, Indiana State University, Terre Haute, IN 47809, USA
| | - Ye Chen
- Department of Mathematics and Statistics, Northern Arizona University, Flagstaff, AZ 86001, USA
| | - Shaad M Ahmad
- Department of Biology, Indiana State University, Terre Haute, IN 47809, USA
- The Center for Genomic Advocacy, Indiana State University, Terre Haute, IN 47809, USA
- Rich and Robin Porter Cancer Research Center, Indiana State University, Terre Haute, IN 47809, USA
| |
Collapse
|
4
|
Chen Y, Zhou X, Wang X, Zhang Y, Song J, Cai Y, Zhao Y, Mei L, Zhu S, Chen X. Fibroblast growth factor 20 ameliorates cardiac hypertrophy via activation ErbB2. Heliyon 2024; 10:e37085. [PMID: 39319165 PMCID: PMC11419851 DOI: 10.1016/j.heliyon.2024.e37085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 08/26/2024] [Accepted: 08/27/2024] [Indexed: 09/26/2024] Open
Abstract
Fibroblast growth factor 20 (FGF20) is a member of the fibroblast growth factor family and involved in embryonic development and cardiac repair. This study aimed to explore the role of FGF20 in cardiac hypertrophy and the underlying molecular mechanisms. FGF20 improved cardiac hypertrophy in vivo and in vitro. Furthermore, FGF20 increased expression of erythroblastic leukemia viral oncogene homolog 2 (ErbB2), which was negatively correlated with expression of the cardiac hypertrophy markers atrial natriuretic peptide (ANP) and brain natriuretic peptide (BNP). In addition, FGF20 effectively protected cardiomyocytes against apoptosis and oxidative stress. To further investigate whether protective effect of FGF20 is mediated by ErbB2, neonatal rat cardiomyocytes (NRCMs) were treated with lapatinib, an inhibitor of ErbB2. Lapatinib largely abrogated the anti-hypertrophic effect of FGF20, accompanied by increases in cardiomyocyte apoptosis and oxidative stress. In summary, this study reveals that FGF20 prevents cardiac hypertrophy by inhibiting apoptosis and oxidative stress via activating ErbB2 and may be a promising therapeutic strategy for cardiac hypertrophy.
Collapse
Affiliation(s)
- Yunjie Chen
- Department of Pharmacy, The First Affiliated Hospital of Ningbo University, Ningbo, 315010, China
| | - Xuan Zhou
- Department of Pharmacy, The First Affiliated Hospital of Ningbo University, Ningbo, 315010, China
| | - Xu Wang
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325035, China
| | - Yuanbin Zhang
- Department of Pharmacy, The First Affiliated Hospital of Ningbo University, Ningbo, 315010, China
| | - Jiayi Song
- Reproductive Medicine Center, The First Affiliated Hospital of Ningbo University, Ningbo, 315010, China
| | - Yan Cai
- Department of Clinical Pharmacy, Ningbo Ninth Hospital, Ningbo, 315020, Zhejiang, China
| | - Yizhuo Zhao
- Department of Pharmacy, The First Affiliated Hospital of Ningbo University, Ningbo, 315010, China
| | - Lin Mei
- Department of Pharmacy, Xiamen Medical College, Xiamen, 361000, Fujian, China
| | - Suyan Zhu
- Department of Pharmacy, The First Affiliated Hospital of Ningbo University, Ningbo, 315010, China
| | - Xueqin Chen
- Department of Traditional Medicine, The First Affiliated Hospital of Ningbo University, Ningbo, 315010, China
| |
Collapse
|
5
|
Pi J, Liu J, Chang H, Chen X, Pan W, Zhang Q, Zhuang T, Liu J, Wang H, Tomlinson B, Chan P, Cheng Y, Yu Z, Zhang L, Zhao Z, Liu Z, Liu J, Zhang Y. Therapeutic efficacy of ECs Foxp1 targeting Hif1α-Hk2 glycolysis signal to restrict angiogenesis. Redox Biol 2024; 75:103281. [PMID: 39083899 PMCID: PMC11342203 DOI: 10.1016/j.redox.2024.103281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Revised: 07/21/2024] [Accepted: 07/21/2024] [Indexed: 08/02/2024] Open
Abstract
Endothelial cells (ECs) rely on glycolysis for energy production to maintain vascular homeostasis and the normalization of hyperglycolysis in tumor vessels has recently gained attention as a therapeutic target. We analyzed the TCGA database and found reduced Foxp1 expression in lung carcinoma. Immunostaining demonstrated reduced expression more restricted at tumor vascular ECs. Therefore, we investigated the function and mechanisms of Foxp1 in EC metabolism for tumor angiogenesis required for tumor growth. EC-Foxp1 deletion mice exhibited a significant increase of tumor and retinal developmental angiogenesis and Hif1α was identified as Foxp1 target gene, and Hk2 as Hif1α target gene. The Foxp1-Hif1α-Hk2 pathway in ECs is important in the regulation of glycolytic metabolism to govern tumor angiogenesis. Finally, we used genetic deletion of EC-Hif1α and RGD-peptide nanoparticles EC target delivery of Hif1α/Hk2-siRNAs to knockdown gene expression which reduced the tumor EC hyperglycolysis state and restricted angiogenesis for tumor growth. This study advances our understanding of EC metabolism for tumor angiogenesis, and meanwhile provides evidence for future therapeutic intervention of hyperglycolysis in tumor ECs for suppression of tumor growth.
Collapse
Affiliation(s)
- Jingjiang Pi
- State Key Laboratory of Cardiovascular Diseases and Medical Innovation Center, Shanghai Heart Failure Research Center, Department of Cardiology, Department of Cardiovascular Surgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China; Shenzhen Ruipuxun Academy for Stem Cell and Regenerative Medicine, Shenzhen, China; Department of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China.
| | - Jie Liu
- State Key Laboratory of Cardiovascular Diseases and Medical Innovation Center, Shanghai Heart Failure Research Center, Department of Cardiology, Department of Cardiovascular Surgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Huan Chang
- Department of Electrophysiology, Jingjiang People's Hospital Affiliated to Yangzhou University, Yangzhou, 225000, China
| | - Xiaoli Chen
- State Key Laboratory of Cardiovascular Diseases and Medical Innovation Center, Shanghai Heart Failure Research Center, Department of Cardiology, Department of Cardiovascular Surgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Wenqi Pan
- State Key Laboratory of Cardiovascular Diseases and Medical Innovation Center, Shanghai Heart Failure Research Center, Department of Cardiology, Department of Cardiovascular Surgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Qi Zhang
- Department of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Tao Zhuang
- State Key Laboratory of Cardiovascular Diseases and Medical Innovation Center, Shanghai Heart Failure Research Center, Department of Cardiology, Department of Cardiovascular Surgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China; Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Jiwen Liu
- State Key Laboratory of Cardiovascular Diseases and Medical Innovation Center, Shanghai Heart Failure Research Center, Department of Cardiology, Department of Cardiovascular Surgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Haikun Wang
- CAS Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Science, University of Chinese Academy of Sciences, 320 Yueyang Rd, Shanghai, 200031, China
| | - Brian Tomlinson
- Faculty of Medicine, Macau University of Science and Technology, Macau SAR, China
| | - Paul Chan
- Division of Cardiology, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Yu Cheng
- Institute for Biomedical Engineering and Nano Science, Tongji University School of Medicine, Shanghai, China
| | - Zuoren Yu
- State Key Laboratory of Cardiovascular Diseases and Medical Innovation Center, Shanghai Heart Failure Research Center, Department of Cardiology, Department of Cardiovascular Surgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Lin Zhang
- State Key Laboratory of Cardiovascular Diseases and Medical Innovation Center, Shanghai Heart Failure Research Center, Department of Cardiology, Department of Cardiovascular Surgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Zhenlin Zhao
- Shenzhen Ruipuxun Academy for Stem Cell and Regenerative Medicine, Shenzhen, China.
| | - Zhongmin Liu
- State Key Laboratory of Cardiovascular Diseases and Medical Innovation Center, Shanghai Heart Failure Research Center, Department of Cardiology, Department of Cardiovascular Surgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China.
| | - Jie Liu
- State Key Laboratory of Cardiovascular Diseases and Medical Innovation Center, Shanghai Heart Failure Research Center, Department of Cardiology, Department of Cardiovascular Surgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China; Shenzhen Ruipuxun Academy for Stem Cell and Regenerative Medicine, Shenzhen, China.
| | - Yuzhen Zhang
- State Key Laboratory of Cardiovascular Diseases and Medical Innovation Center, Shanghai Heart Failure Research Center, Department of Cardiology, Department of Cardiovascular Surgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China.
| |
Collapse
|
6
|
Van Greenen JD, Hockman D. FGF20. Differentiation 2024; 139:100737. [PMID: 38007375 DOI: 10.1016/j.diff.2023.10.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 09/25/2023] [Accepted: 10/24/2023] [Indexed: 11/27/2023]
Abstract
Fibroblast growth factor 20 (FGF20) is a neurotrophic factor and a member of the FGF9 subfamily. It was first identified in Xenopus embryos and was isolated shortly thereafter from the adult rat brain. Its receptors include FGFR4, FGFR3b, FGFR2b and the FGFRc splice forms. In adults it is highly expressed in the brain, while it is expressed in a variety of regions during embryonic development, including the inner ear, heart, hair placodes, mammary buds, dental epithelium and limbs. As a result of its wide-spread expression, FGF20 mouse mutants exhibit a variety of phenotypes including congenital deafness, lack of hair, small kidneys and delayed mammary ductal outgrowth. FGF20 is also associated with human diseases including Parkinson's Disease, cancer and hereditary deafness.
Collapse
Affiliation(s)
- Justine D Van Greenen
- Division of Cell Biology, Department of Human Biology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa; Neuroscience Institute, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Dorit Hockman
- Division of Cell Biology, Department of Human Biology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa; Neuroscience Institute, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa.
| |
Collapse
|
7
|
Ortiz A, Ayhan F, Harper M, Konopka G. Cell type specific roles of FOXP1 during early neocortical murine development. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.08.598089. [PMID: 38895440 PMCID: PMC11185780 DOI: 10.1101/2024.06.08.598089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Cortical development is a tightly controlled process and any deviation during development may increase the susceptibility to neurodevelopmental disorders, such as autism spectrum disorders (ASD). Numerous studies identified mutations in FOXP1, a transcription factor enriched in the neocortex, as causal for ASD and FOXP1 syndrome. Our group has shown that Foxp1 deletion in the mouse cortex leads to overall reduced cortex thickness, alterations in cortical lamination, and changes in the relative thickness of cortical layers. However, the developmental and cell type-specific mechanisms underlying these changes remained unclear. This work characterizes the developmental requirement of neocortical Foxp1 at key embryonic and perinatal ages using a conditional knock-out of Foxp1. We find that Foxp1 deletion results in accelerated pseudo-age during early neurogenesis, increased cell cycle exit during late neurogenesis, altered gene expression and chromatin accessibility, and selective migration deficits in a subset of upper-layer neurons. These data explain the postnatal differences observed in cortical layers and relative cortical thickness. We also highlight genes regulated by FOXP1 and their enrichment with high-confidence ASD or synaptic genes. Together, these results underscore a network of neurodevelopmental disorder-related genes that may serve as potential modulatory targets for postnatal modification relevant to ASD and FOXP1 syndrome.
Collapse
Affiliation(s)
- Ana Ortiz
- Department of Neuroscience, Peter O’Donnell Jr. Brain Institute, UT Southwestern Medical Center, Dallas, TX, USA
| | - Fatma Ayhan
- Department of Neuroscience, Peter O’Donnell Jr. Brain Institute, UT Southwestern Medical Center, Dallas, TX, USA
| | - Matthew Harper
- Department of Neuroscience, Peter O’Donnell Jr. Brain Institute, UT Southwestern Medical Center, Dallas, TX, USA
| | - Genevieve Konopka
- Department of Neuroscience, Peter O’Donnell Jr. Brain Institute, UT Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
8
|
Buth JE, Dyevich CE, Rubin A, Wang C, Gao L, Marks T, Harrison MR, Kong JH, Ross ME, Novitch BG, Pearson CA. Foxp1 suppresses cortical angiogenesis and attenuates HIF-1alpha signaling to promote neural progenitor cell maintenance. EMBO Rep 2024; 25:2202-2219. [PMID: 38600346 PMCID: PMC11094073 DOI: 10.1038/s44319-024-00131-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 03/16/2024] [Accepted: 03/22/2024] [Indexed: 04/12/2024] Open
Abstract
Neural progenitor cells within the cerebral cortex undergo a characteristic switch between symmetric self-renewing cell divisions early in development and asymmetric neurogenic divisions later. Yet, the mechanisms controlling this transition remain unclear. Previous work has shown that early but not late neural progenitor cells (NPCs) endogenously express the autism-linked transcription factor Foxp1, and both loss and gain of Foxp1 function can alter NPC activity and fate choices. Here, we show that premature loss of Foxp1 upregulates transcriptional programs regulating angiogenesis, glycolysis, and cellular responses to hypoxia. These changes coincide with a premature destabilization of HIF-1α, an elevation in HIF-1α target genes, including Vegfa in NPCs, and precocious vascular network development. In vitro experiments demonstrate that stabilization of HIF-1α in Foxp1-deficient NPCs rescues the premature differentiation phenotype and restores NPC maintenance. Our data indicate that the endogenous decline in Foxp1 expression activates the HIF-1α transcriptional program leading to changes in the tissue environment adjacent to NPCs, which, in turn, might alter their self-renewal and neurogenic capacities.
Collapse
Affiliation(s)
- Jessie E Buth
- Department of Neurobiology, Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, Intellectual and Developmental Disabilities Research Center, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
| | - Catherine E Dyevich
- Feil Family Brain and Mind Research Institute and Center for Neurogenetics, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Alexandra Rubin
- Feil Family Brain and Mind Research Institute and Center for Neurogenetics, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Chengbing Wang
- Feil Family Brain and Mind Research Institute and Center for Neurogenetics, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Lei Gao
- Department of Biochemistry, University of Washington, Seattle, WA, 98195, USA
| | - Tessa Marks
- Department of Biochemistry, University of Washington, Seattle, WA, 98195, USA
| | - Michael Rm Harrison
- Cardiovascular Research Institute, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Jennifer H Kong
- Department of Biochemistry, University of Washington, Seattle, WA, 98195, USA
| | - M Elizabeth Ross
- Feil Family Brain and Mind Research Institute and Center for Neurogenetics, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Bennett G Novitch
- Department of Neurobiology, Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, Intellectual and Developmental Disabilities Research Center, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
| | - Caroline Alayne Pearson
- Feil Family Brain and Mind Research Institute and Center for Neurogenetics, Weill Cornell Medicine, New York, NY, 10021, USA.
| |
Collapse
|
9
|
Wu ST, Feng Y, Song R, Qi Y, Li L, Lu D, Wang Y, Wu W, Morgan A, Wang X, Xia Y, Liu R, Alexander SI, Wong J, Zhang Y, Zheng X. Foxp1 Is Required for Renal Intercalated Cell Differentiation and Acid-Base Regulation. J Am Soc Nephrol 2024; 35:533-548. [PMID: 38332484 PMCID: PMC11149051 DOI: 10.1681/asn.0000000000000319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 02/05/2024] [Indexed: 02/10/2024] Open
Abstract
Key Points Foxp1 is a key transcriptional factor for the differentiation of intercalated cells in collecting ducts. Dmrt2 and Hmx2 act downstream of Foxp1 to control the differentiation of type A and type B intercalated cells, respectively. Foxp1 and Dmrt2 are essential for body acid–base balance regulation. Background Kidney collecting ducts comprise principal cells and intercalated cells, with intercalated cells playing a crucial role in kidney acid–base regulation through H+ and HCO3− secretion. Despite its significance, the molecular mechanisms controlling intercalated cell development remain incompletely understood. Methods To investigate the specific role of Foxp1 in kidney tubular system, we specifically deleted Foxp1 expression in kidney distal nephrons and collecting ducts. We examined the effects of Foxp1 on intercalated cell differentiation and urine acidification. RNA sequencing and Chip-seq were used to identify Foxp1 target genes. To dissect the genetic network that regulates intercalated cell differentiation, Dmrt2 -deficient mice were generated to determine the role of Dmrt2 in intercalated cell differentiation. Foxp1 -deficient mice were crossed with Notch2 -deficient mice to dissect the relation between Foxp1 and Notch signaling. Results Foxp1 was selectively expressed in intercalated cells in collecting ducts. The absence of Foxp1 in kidney tubules led to the abolishment of intercalated cell differentiation in the collecting ducts, resulting in distal renal tubular acidosis. Foxp1 regulates the expression of Dmrt2 and Hmx2 , two genes encoding transcription factors specifically expressed in type A and type B intercalated cell cells, respectively. Further genetic analysis revealed that Dmrt2 was essential for type A intercalated cell differentiation, and Foxp1 was necessary downstream of Notch for the regulation of intercalated cell differentiation. Conclusions Foxp1 is required for the renal intercalated cell differentiation and participated in acid–base regulation. Foxp1 regulated downstream transcriptional factors, Dmrt2 and Hmx2, which were involved in the specification of distinct subsets of intercalated cells.
Collapse
Affiliation(s)
- Shi-Ting Wu
- Department of Pharmacology and Tianjin Key Laboratory of Inflammation Biology, School of Basic Medical Sciences, and Center for Cardiovascular Diseases, Tianjin Medical University, Tianjin, China
| | - Yu Feng
- Department of Pharmacology and Tianjin Key Laboratory of Inflammation Biology, School of Basic Medical Sciences, and Center for Cardiovascular Diseases, Tianjin Medical University, Tianjin, China
| | - Renhua Song
- Epigenetics and RNA Biology Program Centenary Institute and the Faulty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Yanmiao Qi
- Department of Pharmacology and Tianjin Key Laboratory of Inflammation Biology, School of Basic Medical Sciences, and Center for Cardiovascular Diseases, Tianjin Medical University, Tianjin, China
| | - Lin Li
- Department of Pharmacology and Tianjin Key Laboratory of Inflammation Biology, School of Basic Medical Sciences, and Center for Cardiovascular Diseases, Tianjin Medical University, Tianjin, China
| | - Dongbo Lu
- Department of Pharmacology and Tianjin Key Laboratory of Inflammation Biology, School of Basic Medical Sciences, and Center for Cardiovascular Diseases, Tianjin Medical University, Tianjin, China
| | - Yixuan Wang
- Department of Pharmacology and Tianjin Key Laboratory of Inflammation Biology, School of Basic Medical Sciences, and Center for Cardiovascular Diseases, Tianjin Medical University, Tianjin, China
| | - Wenrun Wu
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Angela Morgan
- Murdoch Children's Research Institute, The Royal Children's Hospital and Department of Audiology and Speech Pathology and Department of Pediatrics, University of Melbourne, Parkville, Victoria, Australia
| | - Xiaohong Wang
- Department of Pharmacology and Tianjin Key Laboratory of Inflammation Biology, School of Basic Medical Sciences, and Center for Cardiovascular Diseases, Tianjin Medical University, Tianjin, China
| | - Yin Xia
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Renjing Liu
- Vascular Epigenetics Laboratory, Victor Chang Cardiac Research Institute and School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Sydney, New South Wales, Australia
| | - Stephen I. Alexander
- Department of Pediatric Nephrology, The Children's Hospital at Westmead and Centre for Kidney Research, Sydney Medical School, University of Sydney, Sydney, New South Wales, Australia
| | - Justin Wong
- Epigenetics and RNA Biology Program Centenary Institute and the Faulty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Yuzhen Zhang
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xiangjian Zheng
- Department of Pharmacology and Tianjin Key Laboratory of Inflammation Biology, School of Basic Medical Sciences, and Center for Cardiovascular Diseases, Tianjin Medical University, Tianjin, China
| |
Collapse
|
10
|
Steiman S, Miyake T, McDermott JC. FoxP1 Represses MEF2A in Striated Muscle. Mol Cell Biol 2024; 44:57-71. [PMID: 38483114 PMCID: PMC10950271 DOI: 10.1080/10985549.2024.2323959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 01/23/2024] [Indexed: 03/19/2024] Open
Abstract
Myocyte enhancer factor 2 (MEF2) proteins are involved in multiple developmental, physiological, and pathological processes in vertebrates. Protein-protein interactions underlie the plethora of biological processes impacted by MEF2A, necessitating a detailed characterization of the MEF2A interactome. A nanobody based affinity-purification/mass spectrometry strategy was employed to achieve this goal. Specifically, the MEF2A protein complexes were captured from myogenic lysates using a GFP-tagged MEF2A protein immobilized with a GBP-nanobody followed by LC-MS/MS proteomic analysis to identify MEF2A interactors. After bioinformatic analysis, we further characterized the interaction of MEF2A with a transcriptional repressor, FOXP1. FOXP1 coprecipitated with MEF2A in proliferating myogenic cells which diminished upon differentiation (myotube formation). Ectopic expression of FOXP1 inhibited MEF2A driven myogenic reporter genes (derived from the creatine kinase muscle and myogenin genes) and delayed induction of endogenous myogenin during differentiation. Conversely, FOXP1 depletion enhanced MEF2A transactivation properties and myogenin expression. The FoxP1:MEF2A interaction is also preserved in cardiomyocytes and FoxP1 depletion enhanced cardiomyocyte hypertrophy. FOXP1 prevented MEF2A phosphorylation and activation by the p38MAPK pathway. Overall, these data implicate FOXP1 in restricting MEF2A function in order to avoid premature differentiation in myogenic progenitors and also to possibly prevent re-activation of embryonic gene expression in cardiomyocyte hypertrophy.
Collapse
Affiliation(s)
- Sydney Steiman
- Department of Biology, York University, Toronto, ON, Canada
- Muscle Health Research Centre (MHRC), York University, Toronto, ON, Canada
- Centre for Research in Biomolecular Interactions (CRBI), York University, Toronto, ON, Canada
| | - Tetsuaki Miyake
- Department of Biology, York University, Toronto, ON, Canada
- Muscle Health Research Centre (MHRC), York University, Toronto, ON, Canada
- Centre for Research in Biomolecular Interactions (CRBI), York University, Toronto, ON, Canada
| | - John C. McDermott
- Department of Biology, York University, Toronto, ON, Canada
- Muscle Health Research Centre (MHRC), York University, Toronto, ON, Canada
- Centre for Research in Biomolecular Interactions (CRBI), York University, Toronto, ON, Canada
| |
Collapse
|
11
|
Cheng YC, Hsieh ML, Lin CJ, Chang CMC, Huang CY, Puntney R, Wu Moy A, Ting CY, Herr Chan DZ, Nicholson MW, Lin PJ, Chen HC, Kim GC, Zhang J, Coonen J, Basu P, Simmons HA, Liu YW, Hacker TA, Kamp TJ, Hsieh PCH. Combined Treatment of Human Induced Pluripotent Stem Cell-Derived Cardiomyocytes and Endothelial Cells Regenerate the Infarcted Heart in Mice and Non-Human Primates. Circulation 2023; 148:1395-1409. [PMID: 37732466 PMCID: PMC10683868 DOI: 10.1161/circulationaha.122.061736] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 08/23/2023] [Indexed: 09/22/2023]
Abstract
BACKGROUND Remuscularization of the mammalian heart can be achieved after cell transplantation of human induced pluripotent stem cell (hiPSC)-derived cardiomyocytes (CMs). However, several hurdles remain before implementation into clinical practice. Poor survival of the implanted cells is related to insufficient vascularization, and the potential for fatal arrhythmogenesis is associated with the fetal cell-like nature of immature CMs. METHODS We generated 3 lines of hiPSC-derived endothelial cells (ECs) and hiPSC-CMs from 3 independent donors and tested hiPSC-CM sarcomeric length, gap junction protein, and calcium-handling ability in coculture with ECs. Next, we examined the therapeutic effect of the cotransplantation of hiPSC-ECs and hiPSC-CMs in nonobese diabetic-severe combined immunodeficiency (NOD-SCID) mice undergoing myocardial infarction (n≥4). Cardiac function was assessed by echocardiography, whereas arrhythmic events were recorded using 3-lead ECGs. We further used healthy non-human primates (n=4) with cell injection to study the cell engraftment, maturation, and integration of transplanted hiPSC-CMs, alone or along with hiPSC-ECs, by histological analysis. Last, we tested the cell therapy in ischemic reperfusion injury in non-human primates (n=4, 3, and 4 for EC+CM, CM, and control, respectively). Cardiac function was evaluated by echocardiography and cardiac MRI, whereas arrhythmic events were monitored by telemetric ECG recorders. Cell engraftment, angiogenesis, and host-graft integration of human grafts were also investigated. RESULTS We demonstrated that human iPSC-ECs promote the maturity and function of hiPSC-CMs in vitro and in vivo. When cocultured with ECs, CMs showed more mature phenotypes in cellular structure and function. In the mouse model, cotransplantation augmented the EC-accompanied vascularization in the grafts, promoted the maturity of CMs at the infarct area, and improved cardiac function after myocardial infarction. Furthermore, in non-human primates, transplantation of ECs and CMs significantly enhanced graft size and vasculature and improved cardiac function after ischemic reperfusion. CONCLUSIONS These results demonstrate the synergistic effect of combining iPSC-derived ECs and CMs for therapy in the postmyocardial infarction heart, enabling a promising strategy toward clinical translation.
Collapse
Affiliation(s)
- Yu-Che Cheng
- Institute of Biomedical Sciences, Academia Sinica, Taiwan (Y.C.C., C.J.L., C.Y.H., C.Y.T., D.Z.H.C., M.W.N., P.J.L., H.C.C., P.C.H.H.)
| | - Marvin L Hsieh
- Model Organisms Research Core, Department of Medicine (M.L.H., C.M.C.C., T.A.H.), University of Wisconsin-Madison
| | - Chen-Ju Lin
- Institute of Biomedical Sciences, Academia Sinica, Taiwan (Y.C.C., C.J.L., C.Y.H., C.Y.T., D.Z.H.C., M.W.N., P.J.L., H.C.C., P.C.H.H.)
| | - Cindy M C Chang
- Model Organisms Research Core, Department of Medicine (M.L.H., C.M.C.C., T.A.H.), University of Wisconsin-Madison
| | - Ching-Ying Huang
- Institute of Biomedical Sciences, Academia Sinica, Taiwan (Y.C.C., C.J.L., C.Y.H., C.Y.T., D.Z.H.C., M.W.N., P.J.L., H.C.C., P.C.H.H.)
| | - Riley Puntney
- Wisconsin National Primate Research Center (R.P., A.W.M., J.C., P.B., H.A.S.), University of Wisconsin-Madison
| | - Amy Wu Moy
- Wisconsin National Primate Research Center (R.P., A.W.M., J.C., P.B., H.A.S.), University of Wisconsin-Madison
| | - Chien-Yu Ting
- Institute of Biomedical Sciences, Academia Sinica, Taiwan (Y.C.C., C.J.L., C.Y.H., C.Y.T., D.Z.H.C., M.W.N., P.J.L., H.C.C., P.C.H.H.)
| | - Darien Zhing Herr Chan
- Institute of Biomedical Sciences, Academia Sinica, Taiwan (Y.C.C., C.J.L., C.Y.H., C.Y.T., D.Z.H.C., M.W.N., P.J.L., H.C.C., P.C.H.H.)
| | - Martin W Nicholson
- Institute of Biomedical Sciences, Academia Sinica, Taiwan (Y.C.C., C.J.L., C.Y.H., C.Y.T., D.Z.H.C., M.W.N., P.J.L., H.C.C., P.C.H.H.)
| | - Po-Ju Lin
- Institute of Biomedical Sciences, Academia Sinica, Taiwan (Y.C.C., C.J.L., C.Y.H., C.Y.T., D.Z.H.C., M.W.N., P.J.L., H.C.C., P.C.H.H.)
| | - Hung-Chih Chen
- Institute of Biomedical Sciences, Academia Sinica, Taiwan (Y.C.C., C.J.L., C.Y.H., C.Y.T., D.Z.H.C., M.W.N., P.J.L., H.C.C., P.C.H.H.)
| | - Gina C Kim
- Department of Medicine and Stem Cell and Regenerative Medicine Center (G.C.K., J.Z., T.J.K., P.C.H.H.), University of Wisconsin-Madison
| | - Jianhua Zhang
- Department of Medicine and Stem Cell and Regenerative Medicine Center (G.C.K., J.Z., T.J.K., P.C.H.H.), University of Wisconsin-Madison
| | - Jennifer Coonen
- Wisconsin National Primate Research Center (R.P., A.W.M., J.C., P.B., H.A.S.), University of Wisconsin-Madison
| | - Puja Basu
- Wisconsin National Primate Research Center (R.P., A.W.M., J.C., P.B., H.A.S.), University of Wisconsin-Madison
| | - Heather A Simmons
- Wisconsin National Primate Research Center (R.P., A.W.M., J.C., P.B., H.A.S.), University of Wisconsin-Madison
| | - Yen-Wen Liu
- Division of Cardiology, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan (Y.W.L.)
| | - Timothy A Hacker
- Model Organisms Research Core, Department of Medicine (M.L.H., C.M.C.C., T.A.H.), University of Wisconsin-Madison
| | - Timothy J Kamp
- Department of Medicine and Stem Cell and Regenerative Medicine Center (G.C.K., J.Z., T.J.K., P.C.H.H.), University of Wisconsin-Madison
| | - Patrick C H Hsieh
- Institute of Biomedical Sciences, Academia Sinica, Taiwan (Y.C.C., C.J.L., C.Y.H., C.Y.T., D.Z.H.C., M.W.N., P.J.L., H.C.C., P.C.H.H.)
- Department of Medicine and Stem Cell and Regenerative Medicine Center (G.C.K., J.Z., T.J.K., P.C.H.H.), University of Wisconsin-Madison
- Institute of Medical Genomics and Proteomics and Institute of Clinical Medicine, National Taiwan University College of Medicine, Taipei, Taiwan (P.C.H.H.)
| |
Collapse
|
12
|
Liu H, Duan R, He X, Qi J, Xing T, Wu Y, Zhou L, Wang L, Shao Y, Zhang F, Zhou H, Gu X, Lin B, Liu Y, Wang Y, Liu Y, Li L, Liang D, Chen YH. Endothelial deletion of PTBP1 disrupts ventricular chamber development. Nat Commun 2023; 14:1796. [PMID: 37002228 PMCID: PMC10066379 DOI: 10.1038/s41467-023-37409-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 03/16/2023] [Indexed: 04/03/2023] Open
Abstract
The growth and maturation of the ventricular chamber require spatiotemporally precise synergy between diverse cell types. Alternative splicing deeply affects the processes. However, the functional properties of alternative splicing in cardiac development are largely unknown. Our study reveals that an alternative splicing factor polypyrimidine tract-binding protein 1 (PTBP1) plays a key role in ventricular chamber morphogenesis. During heart development, PTBP1 colocalizes with endothelial cells but is almost undetectable in cardiomyocytes. The endothelial-specific knockout of Ptbp1, in either endocardial cells or pan-endothelial cells, leads to a typical phenotype of left ventricular noncompaction (LVNC). Mechanistically, the deletion of Ptbp1 reduces the migration of endothelial cells, disrupting cardiomyocyte proliferation and ultimately leading to the LVNC. Further study shows that Ptbp1 deficiency changes the alternative splicing of β-arrestin-1 (Arrb1), which affects endothelial cell migration. In conclusion, as an alternative splicing factor, PTBP1 is essential during ventricular chamber development, and its deficiency can lead to congenital heart disease.
Collapse
Affiliation(s)
- Hongyu Liu
- Department of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, 200120, Shanghai, China
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Shanghai East Hospital, Tongji University School of Medicine, 200120, Shanghai, China
| | - Ran Duan
- Department of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, 200120, Shanghai, China
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Shanghai East Hospital, Tongji University School of Medicine, 200120, Shanghai, China
| | - Xiaoyu He
- Department of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, 200120, Shanghai, China
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Shanghai East Hospital, Tongji University School of Medicine, 200120, Shanghai, China
| | - Jincu Qi
- Department of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, 200120, Shanghai, China
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Shanghai East Hospital, Tongji University School of Medicine, 200120, Shanghai, China
| | - Tianming Xing
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Shanghai East Hospital, Tongji University School of Medicine, 200120, Shanghai, China
- Jinzhou Medical University, 121000, Jinzhou, Liaoning, China
| | - Yahan Wu
- Department of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, 200120, Shanghai, China
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Shanghai East Hospital, Tongji University School of Medicine, 200120, Shanghai, China
| | - Liping Zhou
- Department of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, 200120, Shanghai, China
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Shanghai East Hospital, Tongji University School of Medicine, 200120, Shanghai, China
| | - Lingling Wang
- Department of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, 200120, Shanghai, China
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Shanghai East Hospital, Tongji University School of Medicine, 200120, Shanghai, China
| | - Yujing Shao
- Department of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, 200120, Shanghai, China
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Shanghai East Hospital, Tongji University School of Medicine, 200120, Shanghai, China
| | - Fulei Zhang
- Department of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, 200120, Shanghai, China
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Shanghai East Hospital, Tongji University School of Medicine, 200120, Shanghai, China
| | - Huixing Zhou
- Department of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, 200120, Shanghai, China
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Shanghai East Hospital, Tongji University School of Medicine, 200120, Shanghai, China
| | - Xingdong Gu
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Shanghai East Hospital, Tongji University School of Medicine, 200120, Shanghai, China
- Jinzhou Medical University, 121000, Jinzhou, Liaoning, China
| | - Bowen Lin
- Department of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, 200120, Shanghai, China
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Shanghai East Hospital, Tongji University School of Medicine, 200120, Shanghai, China
| | - Yuanyuan Liu
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Shanghai East Hospital, Tongji University School of Medicine, 200120, Shanghai, China
- Jinzhou Medical University, 121000, Jinzhou, Liaoning, China
| | - Yan Wang
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Shanghai East Hospital, Tongji University School of Medicine, 200120, Shanghai, China
- Jinzhou Medical University, 121000, Jinzhou, Liaoning, China
| | - Yi Liu
- Department of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, 200120, Shanghai, China
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Shanghai East Hospital, Tongji University School of Medicine, 200120, Shanghai, China
| | - Li Li
- Department of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, 200120, Shanghai, China
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Shanghai East Hospital, Tongji University School of Medicine, 200120, Shanghai, China
- Research Units of Origin and Regulation of Heart Rhythm, Chinese Academy of Medical Sciences, 200092, Shanghai, China
- Department of Pathology and Pathophysiology, Tongji University School of Medicine, 200092, Shanghai, China
| | - Dandan Liang
- Department of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, 200120, Shanghai, China.
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Shanghai East Hospital, Tongji University School of Medicine, 200120, Shanghai, China.
- Research Units of Origin and Regulation of Heart Rhythm, Chinese Academy of Medical Sciences, 200092, Shanghai, China.
| | - Yi-Han Chen
- Department of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, 200120, Shanghai, China.
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Shanghai East Hospital, Tongji University School of Medicine, 200120, Shanghai, China.
- Research Units of Origin and Regulation of Heart Rhythm, Chinese Academy of Medical Sciences, 200092, Shanghai, China.
- Department of Pathology and Pathophysiology, Tongji University School of Medicine, 200092, Shanghai, China.
| |
Collapse
|
13
|
Wakabayashi T, Naito H. Cellular heterogeneity and stem cells of vascular endothelial cells in blood vessel formation and homeostasis: Insights from single-cell RNA sequencing. Front Cell Dev Biol 2023; 11:1146399. [PMID: 37025170 PMCID: PMC10070846 DOI: 10.3389/fcell.2023.1146399] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 03/06/2023] [Indexed: 04/08/2023] Open
Abstract
Vascular endothelial cells (ECs) that constitute the inner surface of blood vessels are essential for new vessel formation and organ homeostasis. ECs display remarkable phenotypic heterogeneity across different organs and the vascular tree during angiogenesis and homeostasis. Recent advances in single cell RNA sequencing (scRNA-seq) technologies have allowed a new understanding of EC heterogeneity in both mice and humans. In particular, scRNA-seq has identified new molecular signatures for arterial, venous and capillary ECs in different organs, as well as previously unrecognized specialized EC subtypes, such as the aerocytes localized in the alveolar capillaries of the lung. scRNA-seq has also revealed the gene expression profiles of specialized tissue-resident EC subtypes that are capable of clonal expansion and contribute to adult angiogenesis, a process of new vessel formation from the pre-existing vasculature. These specialized tissue-resident ECs have been identified in various different mouse tissues, including aortic endothelium, liver, heart, lung, skin, skeletal muscle, retina, choroid, and brain. Transcription factors and signaling pathways have also been identified in the specialized tissue-resident ECs that control angiogenesis. Furthermore, scRNA-seq has also documented responses of ECs in diseases such as cancer, age-related macular degeneration, Alzheimer's disease, atherosclerosis, and myocardial infarction. These new findings revealed by scRNA-seq have the potential to provide new therapeutic targets for different diseases associated with blood vessels. In this article, we summarize recent advances in the understanding of the vascular endothelial cell heterogeneity and endothelial stem cells associated with angiogenesis and homeostasis in mice and humans, and we discuss future prospects for the application of scRNA-seq technology.
Collapse
Affiliation(s)
- Taku Wakabayashi
- Department of Ophthalmology, Osaka University Graduate School of Medicine, Osaka, Japan
- Wills Eye Hospital, Thomas Jefferson University, Philadelphia, PA, United States
- *Correspondence: Taku Wakabayashi, ; Hisamichi Naito,
| | - Hisamichi Naito
- Department of Vascular Physiology, Kanazawa University Graduate School of Medical Science, Kanazawa, Ishikawa, Japan
- *Correspondence: Taku Wakabayashi, ; Hisamichi Naito,
| |
Collapse
|
14
|
Transcription factor Foxp1 stimulates angiogenesis in adult rats after myocardial infarction. Cell Death Dis 2022; 8:381. [PMID: 36088337 PMCID: PMC9464245 DOI: 10.1038/s41420-022-01180-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 09/01/2022] [Accepted: 09/02/2022] [Indexed: 11/15/2022]
Abstract
Forkhead box protein P1 (FoxP1) is essential for cardiac development and the regulation of neovascularization, but its potential for cardiac angiogenesis has not been explored. This study aims to investigate the angiogenic role of FoxP1 in a rat model of myocardial infarction (MI). Adult male rats were subjected to MI, and Foxp1 was knocked down with lentivirus FoxP1 siRNA. Endothelial cell proliferation, angiogenesis, and cardiac function were also assessed. Cell scratch assay and tubule formation analysis were used to detect the migration ability and tube formation ability of human umbilical vein endothelial cells (HUVECs). Compared with that in the sham group, results showed that the expression of FoxP1 was significantly increased in the MI group. Foxp1 knockdown decreases FoxP1 expression, reduces angiogenesis, and increases collagen deposition. When Foxp1 was knocked down in HUVECs using FoxP1 siRNA lentivirus, cell proliferation, migration, and tube formation abilities decreased significantly. Our study showed that FoxP1 elicits pleiotropic beneficial actions on angiogenesis in the post-MI heart by promoting the proliferation of endothelial cells. FoxP1 should be considered a candidate for therapeutic cardiac angiogenesis.
Collapse
|
15
|
Chen X, Xu J, Bao W, Li H, Wu W, Liu J, Pi J, Tomlinson B, Chan P, Ruan C, Zhang Q, Zhang L, Fan H, Morrisey E, Liu Z, Zhang Y, Lin L, Liu J, Zhuang T. Endothelial Foxp1 Regulates Neointimal Hyperplasia Via Matrix Metalloproteinase-9/Cyclin Dependent Kinase Inhibitor 1B Signal Pathway. J Am Heart Assoc 2022; 11:e026378. [PMID: 35904197 PMCID: PMC9375493 DOI: 10.1161/jaha.122.026378] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background The endothelium is essential for maintaining vascular physiological homeostasis and the endothelial injury leads to the neointimal hyperplasia because of the excessive proliferation of vascular smooth muscle cells. Endothelial Foxp1 (forkhead box P1) has been shown to control endothelial cell (EC) proliferation and migration in vitro. However, whether EC-Foxp1 participates in neointimal formation in vivo is not clear. Our study aimed to investigate the roles and mechanisms of EC-Foxp1 in neointimal hyperplasia. Methods and Results The wire injury femoral artery neointimal hyperplasia model was performed in Foxp1 EC-specific loss-of-function and gain-of-function mice. EC-Foxp1 deletion mice displayed the increased neointimal formation through elevation of vascular smooth muscle cell proliferation and migration, and the reduction of EC proliferation hence reendothelialization after injury. In contrast, EC-Foxp1 overexpression inhibited the neointimal formation. EC-Foxp1 paracrine regulated vascular smooth muscle cell proliferation and migration via targeting matrix metalloproteinase-9. Also, EC-Foxp1 deletion impaired EC repair through reduction of EC proliferation via increasing cyclin dependent kinase inhibitor 1B expression. Delivery of cyclin dependent kinase inhibitor 1B-siRNA to ECs using RGD (Arg-Gly-Asp)-peptide magnetic nanoparticle normalized the EC-Foxp1 deletion-mediated impaired EC repair and attenuated the neointimal formation. EC-Foxp1 regulates matrix metalloproteinase-9/cyclin dependent kinase inhibitor 1B signaling pathway to control injury induced neointimal formation. Conclusions Our study reveals that targeting EC-Foxp1-matrix metalloproteinase-9/cyclin dependent kinase inhibitor 1B pathway might provide future novel therapeutic interventions for restenosis.
Collapse
Affiliation(s)
- Xiaoli Chen
- Key Laboratory of Arrhythmias of the Ministry of Education of ChinaResearch Center for Translational MedicineShanghai East HospitalTongji University School of MedicineShanghaiChina
| | - Jianfei Xu
- Key Laboratory of Arrhythmias of the Ministry of Education of ChinaResearch Center for Translational MedicineShanghai East HospitalTongji University School of MedicineShanghaiChina
| | - Wenzhen Bao
- Key Laboratory of Arrhythmias of the Ministry of Education of ChinaResearch Center for Translational MedicineShanghai East HospitalTongji University School of MedicineShanghaiChina
| | - Hongda Li
- Key Laboratory of Arrhythmias of the Ministry of Education of ChinaResearch Center for Translational MedicineShanghai East HospitalTongji University School of MedicineShanghaiChina
| | - Wenrun Wu
- Key Laboratory of Arrhythmias of the Ministry of Education of ChinaResearch Center for Translational MedicineShanghai East HospitalTongji University School of MedicineShanghaiChina
| | - Jiwen Liu
- Key Laboratory of Arrhythmias of the Ministry of Education of ChinaResearch Center for Translational MedicineShanghai East HospitalTongji University School of MedicineShanghaiChina
| | - Jingjiang Pi
- Department of CardiologyShanghai East HospitalTongji University School of MedicineShanghaiChina
| | - Brian Tomlinson
- Faculty of MedicineMacau University of Science and TechnologyMacauChina
| | - Paul Chan
- Division of CardiologyDepartment of Internal MedicineWan Fang HospitalTaipei Medical UniversityTaipeiTaiwan
| | - Chengchao Ruan
- Department of Physiology and Pathophysiology School of Basic Medical SciencesFudan UniversityShanghaiChina
| | - Qi Zhang
- Department of CardiologyShanghai East HospitalTongji University School of MedicineShanghaiChina
| | - Lin Zhang
- Key Laboratory of Arrhythmias of the Ministry of Education of ChinaResearch Center for Translational MedicineShanghai East HospitalTongji University School of MedicineShanghaiChina
| | - Huimin Fan
- Key Laboratory of Arrhythmias of the Ministry of Education of ChinaResearch Center for Translational MedicineShanghai East HospitalTongji University School of MedicineShanghaiChina
| | - Edward Morrisey
- Department of Cell and Developmental Biology (R.W., E.E.M.)Department of Medicine (E.E.M.)Penn Cardiovascular Institute (E.E.M.), and Penn Institute for Regenerative Medicine (E.E.M.)University of PennsylvaniaPhiladelphiaPennsylvania
| | - Zhongmin Liu
- Key Laboratory of Arrhythmias of the Ministry of Education of ChinaResearch Center for Translational MedicineShanghai East HospitalTongji University School of MedicineShanghaiChina
| | - Yuzhen Zhang
- Key Laboratory of Arrhythmias of the Ministry of Education of ChinaResearch Center for Translational MedicineShanghai East HospitalTongji University School of MedicineShanghaiChina
| | - Li Lin
- Department of CardiologyShanghai East HospitalTongji University School of MedicineShanghaiChina
| | - Jie Liu
- Key Laboratory of Arrhythmias of the Ministry of Education of ChinaResearch Center for Translational MedicineShanghai East HospitalTongji University School of MedicineShanghaiChina
| | - Tao Zhuang
- Key Laboratory of Arrhythmias of the Ministry of Education of ChinaResearch Center for Translational MedicineShanghai East HospitalTongji University School of MedicineShanghaiChina,Department of Physiology and Pathophysiology School of Basic Medical SciencesFudan UniversityShanghaiChina,Shanghai Jinshan Eye Disease Prevention and Treatment InstituteShanghai Jinshan Nuclear and Chemical Injury Emergency Treatment CenterJinshan HospitalFudan UniversityShanghaiChina
| |
Collapse
|
16
|
Chen Y, An N, Zhou X, Mei L, Sui Y, Chen G, Chen H, He S, Jin C, Hu Z, Li W, Wang Y, Lin Z, Chen P, Jin L, Guan X, Wang X. Fibroblast growth factor 20 attenuates pathological cardiac hypertrophy by activating the SIRT1 signaling pathway. Cell Death Dis 2022; 13:276. [PMID: 35351862 PMCID: PMC8964679 DOI: 10.1038/s41419-022-04724-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 02/17/2022] [Accepted: 03/11/2022] [Indexed: 11/08/2022]
Abstract
Cardiac hypertrophy occurs initially in response to an increased cardiac load as a compensatory mechanism to maintain cardiac output. However, sustained pathological hypertrophy can develop into heart failure and cause sudden death. Fibroblast growth factor 20 (FGF20) is a member of the fibroblast growth factor family, which involved in apoptosis, aging, inflammation, and autophagy. The precise function of FGF20 in pathological cardiac hypertrophy is unclear. In this study, we demonstrated that FGF20 was significantly decreased in response to hypertrophic stimulation. In contrast, overexpression of FGF20 protected against pressure overload-induced cardiac hypertrophy. Mechanistically, we found that FGF20 upregulates SIRT1 expression, causing deacetylation of FOXO1; this effect promotes the transcription of downstream antioxidant genes, thus inhibits oxidative stress. In content, the anti-hypertrophic effect of FGF20 was largely counteracted in SIRT1-knockout mice, accompanied by an increase in oxidative stress. In summary, our findings reveal a previously unknown protective effect of FGF20 on pathological cardiac hypertrophy by reducing oxidative stress through activation of the SIRT1 signaling pathway. FGF20 is a potential novel molecular target for preventing and treating pressure overload-induced myocardial injury.
Collapse
Affiliation(s)
- Yunjie Chen
- School of Pharmaceutical Science, Wenzhou Medical University, 325000, Wenzhou, PR China
- Department of Pharmacy, Ningbo first Hospital, 315010, Ningbo, PR China
| | - Ning An
- Department of Pharmacy, Ningbo Medical Center Lihuili Hospital, 315041, Ningbo, PR China
| | - Xuan Zhou
- Department of Pharmacy, Ningbo first Hospital, 315010, Ningbo, PR China
| | - Lin Mei
- School of Pharmaceutical Science, Wenzhou Medical University, 325000, Wenzhou, PR China
| | - Yanru Sui
- School of Pharmaceutical Science, Wenzhou Medical University, 325000, Wenzhou, PR China
| | - Gen Chen
- School of Pharmaceutical Science, Wenzhou Medical University, 325000, Wenzhou, PR China
| | - Huinan Chen
- School of Pharmaceutical Science, Wenzhou Medical University, 325000, Wenzhou, PR China
| | - Shengqu He
- School of Pharmaceutical Science, Wenzhou Medical University, 325000, Wenzhou, PR China
| | - Cheng Jin
- School of Pharmaceutical Science, Wenzhou Medical University, 325000, Wenzhou, PR China
| | - Zhicheng Hu
- School of Pharmaceutical Science, Wenzhou Medical University, 325000, Wenzhou, PR China
| | - Wanqian Li
- School of Pharmaceutical Science, Wenzhou Medical University, 325000, Wenzhou, PR China
| | - Yang Wang
- School of Pharmaceutical Science, Wenzhou Medical University, 325000, Wenzhou, PR China
| | - Zhu Lin
- Department of Pharmacy, Ningbo first Hospital, 315010, Ningbo, PR China
| | - Peng Chen
- Department of Cardiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, 325000, Wenzhou, PR China
| | - Litai Jin
- School of Pharmaceutical Science, Wenzhou Medical University, 325000, Wenzhou, PR China.
| | - Xueqiang Guan
- Department of Cardiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, 325000, Wenzhou, PR China.
| | - Xu Wang
- School of Pharmaceutical Science, Wenzhou Medical University, 325000, Wenzhou, PR China.
| |
Collapse
|
17
|
Gao F, Zhao Y, Zhang B, Xiao C, Sun Z, Gao Y, Dou X. Forkhead box protein 1 transcriptionally activates sestrin1 to alleviate oxidized low-density lipoprotein-induced inflammation and lipid accumulation in macrophages. Bioengineered 2022; 13:2917-2926. [PMID: 35043753 PMCID: PMC8974195 DOI: 10.1080/21655979.2021.2000228] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Transcription factor forkhead box protein 1 (FOXP1) has been shown cardiovascular protection. We aimed to analyze the role of FOXP1 in oxidized low-density lipoprotein (ox-LDL)-induced macrophages and its possible regulatory effect on sestrin1 (SESN1) expression. After stimulation with ox-LDL, FOXP1 expression in RAW264.7 cells was evaluated with RT-qPCR and Western blotting. Then, FOXP1 was overexpressed, followed by detection of inflammatory mediator levels using ELISA kits and RT-qPCR. Lipid accumulation was detected with oil red O staining. Additionally, the JASPAR database was used to predict the potential genes that could be transcriptionally regulated by FOXP1. ChIP and luciferase reporter assays were used to verify this combination. To further clarify the regulatory effects of FOXP1 on SESN1 in damage of macrophages triggered by ox-LDL, SESN1 was silenced to determine the inflammation and lipid accumulation under the condition of FOXP1 overexpression. Results indicated that ox-LDL stimulation led to a significant decrease in FOXP1 expression. FOXP1 overexpression notably reduced the levels of tumor necrosis factor (TNF)-α, interleukin (IL)-1β and IL-6, accompanied by a decreased in phosphorylated NF-κB p65 expression. Besides, FOXP1-upregulation inhibited lipid accumulation and reduced CD36 expression level in RAW264.7 cells upon ox-LDL stimulation. Moreover, results of ChIP and luciferase reporter assays suggested that FOXP1 could transcriptionally regulate SESN1 expression. Further experiments supported that SESN1 silencing restored the inhibitory effects of FOXP1 overexpression on the inflammation and lipid accumulation in RAW264.7 cells exposed to ox-LDL. Collectively, FOXP1 transcriptionally activates SESN1 for the alleviation of ox-LDL-induced inflammation and lipid accumulation in macrophages.
Collapse
Affiliation(s)
- Feng Gao
- Department of Cardiovascular Surgery, Xuzhou Cancer Hospital, Xuzhou, People's Republic of China
| | - Yongcheng Zhao
- Department of Cardiovascular Surgery, Xuzhou Cancer Hospital, Xuzhou, People's Republic of China
| | - Bin Zhang
- Department of Cardiovascular Surgery, Xuzhou Cancer Hospital, Xuzhou, People's Republic of China
| | - Chunwei Xiao
- Department of Cardiovascular Surgery, Xuzhou Cancer Hospital, Xuzhou, People's Republic of China
| | - Zhanfa Sun
- Department of Cardiovascular Surgery, Xuzhou Cancer Hospital, Xuzhou, People's Republic of China
| | - Yuan Gao
- Department of Cardiovascular Surgery, Xuzhou Cancer Hospital, Xuzhou, People's Republic of China
| | - Xueyong Dou
- Department of Cardiovascular Surgery, Xuzhou Cancer Hospital, Xuzhou, People's Republic of China
| |
Collapse
|
18
|
Liu X, Yang Y, Song J, Li D, Liu X, Li C, Ma Z, Zhong J, Wang L. Knockdown of forkhead box protein P1 alleviates hypoxia reoxygenation injury in H9c2 cells through regulating Pik3ip1/Akt/eNOS and ROS/mPTP pathway. Bioengineered 2022; 13:1320-1334. [PMID: 35000528 PMCID: PMC8805992 DOI: 10.1080/21655979.2021.2016046] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Forkhead box protein P1 (Foxp1) exerts an extensive array of physiological and pathophysiological impacts on the cardiovascular system. However, the exact function of myocardial Foxp1 in myocardial ischemic reperfusion injury (MIRI) stays largely vague. The hypoxia reoxygenation model of H9c2 cells (the rat ventricular myoblasts) closely mimics myocardial ischemia-reperfusion injury. This report intends to research the effects and mechanisms underlying Foxp1 on H9c2 cells in response to hypoxia (12 h)/reoxygenation (4 h) (HR) stimulation. Expressions of Foxp1 and Phosphatidylinositol 3-kinase interacting protein 1 (Pik3ip1) were both upregulated in ischemia/reperfusion (IR)/HR-induced injury. Stimulation through HR led to marked increases in cellular apoptosis, mitochondrial dysfunction, and superoxide generation in H9c2 cells, which were rescued with knockdown of Foxp1 by siRNA. Silence of Foxp1 depressed expression of Pik3ip1 directly activated the PI3K/Akt/eNOS pathway and promoted nitric oxide (NO) release. Moreover, the knockdown of Foxp1 blunted HR-induced enhancement of reactive oxygen species (ROS) generation, thus alleviating excessive persistence of mitochondrial permeability transition pore (mPTP) opening and decreased mitochondrial apoptosis-associated protein expressions in H9c2 cells. Meanwhile, these cardioprotective effects can be abolished by LY294002, NG-nitro-L-arginine methyl ester (L-NAME), and Atractyloside (ATR), respectively. In summary, our findings indicated that knockdown of Foxp1 prevented HR-induced encouragement of apoptosis and oxidative stress via PI3K/Akt/eNOS signaling activation by targeting Pik3ip1 and improved mitochondrial function by inhibiting ROS-mediated mPTP opening. Inhibition of Foxp1 may be a promising therapeutic avenue for MIRI.
Collapse
Affiliation(s)
- Xinming Liu
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Yixing Yang
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Jiawei Song
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Dongjie Li
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Xiaoyan Liu
- Medical Research Center, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Chuang Li
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Zheng Ma
- Department of Cardiology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Jiuchang Zhong
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China.,Beijing Institute of Respiratory Medicine, Beijing, China
| | - Lefeng Wang
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
19
|
Hu J, Liu X, Tang Y. HMGB1/Foxp1 regulates hypoxia-induced inflammatory response in macrophages. Cell Biol Int 2021; 46:265-277. [PMID: 34816539 DOI: 10.1002/cbin.11728] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 10/14/2021] [Accepted: 11/13/2021] [Indexed: 01/04/2023]
Abstract
Forkhead box protein P1 (Foxp1) is a kind of tumor suppressor gene, and the role of Foxp1 in the macrophages of myocardial infarction (MI) has not been studied yet. Here, we verified the role of the transcription factor high mobility group box 1 (HMGB1) and its target gene Foxp1 in the inflammatory response. In this study, the key genes HMGB1 and Foxp1 in the macrophages of mouse MI model were screened out through single-cell transcriptome analysis of GSE136088 (GEO database). In vitro experiment indicated that hypoxia induced the inflammatory response in RAW264.7 macrophages, promoted the secretion of inflammatory factors (tumor necrosis factor α [TNF-α], interleukin 6 [IL-6], and IL-1β) and the activation of NLRP3 inflammasome (NLRP3, ASC, and pro-caspase-1). Meanwhile, HMGB1 increased while Foxp1 decreased in hypoxia-treated RAW264.7 macrophages. HMGB1 bound to the upstream promoter region of Foxp1 as demonstrated by the dual-luciferase reporter assay, chromatin immunoprecipitation (ChIP)-quantitative polymerase chain reaction (qPCR) and agarose gel electrophoresis. As a transcription factor, HMGB1 regulated Foxp1 expression. The secretion of inflammatory factors and the expression of NLRP3 inflammasome protein were changed when the expression of HMGB1 and Foxp1 was regulated in the hypoxia-treated RAW264.7 macrophages. This study verified that HMGB1 could aggravate the hypoxia-treated inflammatory response of macrophages through downregulating Foxp1, which not only provides evidence to support the role of HMGB1/Foxp1 in macrophages but also offers another angle for the treatment of MI.
Collapse
Affiliation(s)
- Jing Hu
- Department of Cardiovascular, The First Hospital of Nanchang, Nanchang, Jiangxi, China
| | - Xiaojun Liu
- Department of Cardiovascular, The First Hospital of Nanchang, Nanchang, Jiangxi, China
| | - Yu Tang
- Department of Cardiovascular, Jiangxi Provincial People's Hospital, Nanchang, Jiangxi, China
| |
Collapse
|
20
|
Motawi TMK, Sabry D, Shehata NI, William MM, Fahim AT. Impact of FOXP1 rs2687201 genetic variant on the susceptibility to HCV-related hepatocellular carcinoma in Egyptians. J Biochem Mol Toxicol 2021; 36:e22965. [PMID: 34783112 DOI: 10.1002/jbt.22965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2020] [Revised: 08/25/2021] [Accepted: 10/26/2021] [Indexed: 12/24/2022]
Abstract
Hepatocellular carcinoma (HCC) constitutes a challenging health problem in Egypt due to the high incidence of hepatitis C virus (HCV) infection. Improved understanding of genetic mechanisms underlying the individual predisposition to HCC will lead to enhancements in the early diagnosis, treatment, and prevention of this disease. Transcription factor forkhead box P1 (FOXP1) is involved in the cellular processes of proliferation, differentiation, metabolism, and longevity. In addition, it has been implicated in hepatic tumorigenesis. The present study explored the association of C/A single-nucleotide polymorphism in the FOXP1 gene (rs2687201) with HCC susceptibility in HCV Egyptian patients. The study included 108 patients with HCV-dependant HCC, 86 HCV patients, and 80- age and gender-matched healthy controls. rs2687201 genotyping was performed by allelic discrimination method using TaqMan real-time PCR assays while FOXP1 gene expression and protein level were determined using qRT-PCR and enzyme-linked immunoassay, respectively. Our results revealed a significant association between FOXP1 rs2687201 and HCC risk where (A) allele was significantly more frequent in patients with HCC compared to controls (odds ratio [OR]: 1.88, 95% confidence interval [CI]: 1.17-3.04, p = 0.01) and to HCV patients (OR: 1.85, 95% CI: 1.62-2.94, p = 0.012). Furthermore, FOXP1 gene and protein expression levels were remarkably higher in (CA + AA) than in CC genotype carriers in a dominant model. The (CA + AA) genotype displayed a significantly shorter overall survival than the CC genotype in HCC patients. In conclusion, FOXP1 gene polymorphism rs2687201 is significantly associated with HCC, but not with HCV infection, in Egyptian patients.
Collapse
Affiliation(s)
| | - Dina Sabry
- Department of Medical Biochemistry and Molecular Biology, Cairo University, Cairo, Egypt
| | | | - Mira Magdy William
- Department of Biochemistry, Faculty of Pharmacy, October 6 University, Cairo, Egypt
| | - Atef Tadros Fahim
- Department of Biochemistry, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| |
Collapse
|
21
|
Hubert F, Payan SM, Pelce E, Bouchard L, Sturny R, Lenfant N, Mottola G, Collart F, Kelly RG, Rochais F. FGF10 promotes cardiac repair through a dual cellular mechanism increasing cardiomyocyte renewal and inhibiting fibrosis. Cardiovasc Res 2021; 118:2625-2637. [PMID: 34755840 DOI: 10.1093/cvr/cvab340] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Accepted: 11/05/2021] [Indexed: 11/13/2022] Open
Abstract
AIMS Promoting cardiomyocyte renewal represents a major therapeutic approach for heart regeneration and repair. Our study aims to investigate the relevance of FGF10 as a potential target for heart regeneration. METHODS AND RESULTS Our results first reveal that Fgf10 levels are upregulated in the injured ventricle after MI. Adult mice with reduced Fgf10 expression subjected to MI display impaired cardiomyocyte proliferation and enhanced cardiac fibrosis, leading to a worsened cardiac function and remodeling post-MI. In contrast, conditional Fgf10 overexpression post-MI revealed that, by enhancing cardiomyocyte proliferation and preventing scar-promoting myofibroblast activation, FGF10 preserves cardiac remodeling and function. Moreover, FGF10 activates major regenerative pathways including the regulation of Meis1 expression levels, the Hippo signaling pathway and a pro-glycolytic metabolic switch. Finally, we demonstrate that elevated FGF10 levels in failing human hearts correlate with reduced fibrosis and enhanced cardiomyocyte proliferation. CONCLUSIONS Altogether, our study shows that FGF10 promotes cardiac regeneration and repair through two cellular mechanisms: elevating cardiomyocyte renewal and limiting fibrosis. This study thus identifies FGF10 as a clinically relevant target for heart regeneration and repair in man.
Collapse
Affiliation(s)
- Fabien Hubert
- Aix Marseille Univ, INSERM, MMG, U 1251, Marseille, France
| | - Sandy M Payan
- Aix Marseille Univ, INSERM, MMG, U 1251, Marseille, France
| | - Edeline Pelce
- Aix Marseille Univ, INSERM, MMG, U 1251, Marseille, France.,Department of Cardiac Surgery, Timone Hospital, AP-HM, Marseille, France
| | | | - Rachel Sturny
- Aix Marseille Univ, CNRS UMR 7288, IBDM, Marseille, France
| | | | - Giovanna Mottola
- Aix-Marseille Univ, C2VN, INSERM 1263, INRAE 1260, Marseille, France.,Laboratory of Biochemistry, Timone Hospital, Marseille, France
| | - Frédéric Collart
- Department of Cardiac Surgery, Timone Hospital, AP-HM, Marseille, France
| | - Robert G Kelly
- Aix Marseille Univ, CNRS UMR 7288, IBDM, Marseille, France
| | | |
Collapse
|
22
|
Zhang W, Liu P, Ling S, Wang F, Wang S, Chen T, Zhou R, Xia X, Yao Z, Fan Y, Wang N, Wang J, Tucker HO, Guo X. Forkhead box P1 (Foxp1) in osteoblasts regulates bone mass accrual and adipose tissue energy metabolism. J Bone Miner Res 2021; 36:2017-2026. [PMID: 34131944 DOI: 10.1002/jbmr.4394] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 06/07/2021] [Accepted: 06/12/2021] [Indexed: 11/08/2022]
Abstract
Adiponectin (AdipoQ), a hormone abundantly secreted by adipose tissues, has multiple beneficial functions, including insulin sensitization as well as lipid and glucose metabolism. It has been reported that bone controls energy metabolism through an endocrine-based mechanism. In this study, we observed that bone also acts as an important endocrine source for AdipoQ, and its capacity in osteoblasts is controlled by the forkhead box P1 (FOXP1) transcriptional factor. Deletion of the Foxp1 gene in osteoblasts led to augmentation of AdipoQ levels accompanied by fueled energy expenditure in adipose tissues. In contrast, overexpression of Foxp1 in bones impaired AdipoQ secretion and restrained energy consumption. Chromatin immunoprecipitation sequencing (ChIP-seq) analysis revealed that AdipoQ expression, which increases as a function of bone age, is directly controlled by FOXP1. Our results indicate that bones, especially aged bones, provide an important source of a set of endocrine factors, including AdipoQ, that control body metabolism. © 2021 American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Wei Zhang
- Department of Nephrology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China.,Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Shanghai jiao Tong University, Shanghai, China
| | - Pei Liu
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Shanghai jiao Tong University, Shanghai, China
| | - Shifeng Ling
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Shanghai jiao Tong University, Shanghai, China
| | - Fuhua Wang
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Shanghai jiao Tong University, Shanghai, China
| | - Shaojiao Wang
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Shanghai jiao Tong University, Shanghai, China
| | - Tienan Chen
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Shanghai jiao Tong University, Shanghai, China
| | - Rujiang Zhou
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Shanghai jiao Tong University, Shanghai, China
| | - Xuechun Xia
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Shanghai jiao Tong University, Shanghai, China
| | - Zhengju Yao
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Shanghai jiao Tong University, Shanghai, China
| | - Ying Fan
- Department of Nephrology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Niansong Wang
- Department of Nephrology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Jiqiu Wang
- Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, China
| | - Haley O Tucker
- Institute for Cellular and Molecular Biology, University of Texas at Austin, Austin, TX, USA
| | - Xizhi Guo
- Department of Nephrology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China.,Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Shanghai jiao Tong University, Shanghai, China
| |
Collapse
|
23
|
Wang W, Lu G, Liu H, Xiong Z, Leung H, Cao R, Pang AL, Su X, Law PWN, Zhao Z, Chen Z, Chan W. Pten Regulates Cardiomyocyte Differentiation by Modulating Non-CG Methylation via Dnmt3. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2100849. [PMID: 34247447 PMCID: PMC8425920 DOI: 10.1002/advs.202100849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 06/04/2021] [Indexed: 06/13/2023]
Abstract
The regulation of cardiomyocyte differentiation is a fundamental aspect of cardiac development and regenerative medicine. PTEN plays important roles during embryonic development. However, its role in cardiomyocyte differentiation remains unknown. In this study, a low-cost protocol for cardiomyocyte differentiation from mouse embryonic stem cells (ESCs) is presented and it is shown that Pten deletion potently suppresses cardiomyocyte differentiation. Transcriptome analysis shows that the expression of a series of cardiomyocyte marker genes is downregulated in Pten-/- cardiomyocytes. Pten ablation induces Dnmt3b expression via the AKT/FoxO3a pathway and regulates the expression of a series of imprinted genes, including Igf2. Double knockout of Dnmt3l and Dnmt3b rescues the deficiency of cardiomyocyte differentiation of Pten-/- ESCs. The DNA methylomes from wild-type and Pten-/- embryoid bodies and cardiomyocytes are analyzed by whole-genome bisulfite sequencing. Pten deletion significantly promotes the non-CG (CHG and CHH) methylation levels of genomic DNA during cardiomyocyte differentiation, and the non-CG methylation levels of cardiomyocyte genes and Igf2 are increased in Pten-/- cardiomyocytes. Igf2 or Igf1r deletion also suppresses cardiomyocyte differentiation through the MAPK/ERK signaling pathway, and IGF2 supplementation partially rescues the cardiomyocyte differentiation. Finally, Pten conditional knockout mice are generated and the role of PTEN in cardiomyocyte differentiation is verified in vivo.
Collapse
Affiliation(s)
- Wuming Wang
- CUHK‐SDU Joint Laboratory on Reproductive GeneticsSchool of Biomedical SciencesThe Chinese University of Hong KongHong KongChina
- National Research Center for Assisted Reproductive Technology and Reproductive GeneticsJinan250001China
| | - Gang Lu
- CUHK‐SDU Joint Laboratory on Reproductive GeneticsSchool of Biomedical SciencesThe Chinese University of Hong KongHong KongChina
- National Research Center for Assisted Reproductive Technology and Reproductive GeneticsJinan250001China
| | - Hong‐Bin Liu
- CUHK‐SDU Joint Laboratory on Reproductive GeneticsSchool of Biomedical SciencesThe Chinese University of Hong KongHong KongChina
- National Research Center for Assisted Reproductive Technology and Reproductive GeneticsJinan250001China
| | - Zhiqiang Xiong
- National Research Center for Assisted Reproductive Technology and Reproductive GeneticsJinan250001China
- SDIVF R&D Centre12W, Hong Kong Science ParkShatinHong KongChina
| | - Ho‐Duen Leung
- SDIVF R&D Centre12W, Hong Kong Science ParkShatinHong KongChina
| | - Ruican Cao
- CUHK‐SDU Joint Laboratory on Reproductive GeneticsSchool of Biomedical SciencesThe Chinese University of Hong KongHong KongChina
- National Research Center for Assisted Reproductive Technology and Reproductive GeneticsJinan250001China
| | - Alan Lap‐Yin Pang
- R&D DivisionTGD Life Company Limited15W, Hong Kong Science ParkShatinHong KongChina
| | - Xianwei Su
- CUHK‐SDU Joint Laboratory on Reproductive GeneticsSchool of Biomedical SciencesThe Chinese University of Hong KongHong KongChina
- National Research Center for Assisted Reproductive Technology and Reproductive GeneticsJinan250001China
- SDIVF R&D Centre12W, Hong Kong Science ParkShatinHong KongChina
| | - Patrick Wai Nok Law
- CUHK‐SDU Joint Laboratory on Reproductive GeneticsSchool of Biomedical SciencesThe Chinese University of Hong KongHong KongChina
| | - Zhiju Zhao
- CUHK‐SDU Joint Laboratory on Reproductive GeneticsSchool of Biomedical SciencesThe Chinese University of Hong KongHong KongChina
| | - Zi‐Jiang Chen
- CUHK‐SDU Joint Laboratory on Reproductive GeneticsSchool of Biomedical SciencesThe Chinese University of Hong KongHong KongChina
- National Research Center for Assisted Reproductive Technology and Reproductive GeneticsJinan250001China
| | - Wai‐Yee Chan
- CUHK‐SDU Joint Laboratory on Reproductive GeneticsSchool of Biomedical SciencesThe Chinese University of Hong KongHong KongChina
- National Research Center for Assisted Reproductive Technology and Reproductive GeneticsJinan250001China
| |
Collapse
|
24
|
Neyroud D, Nosacka RL, Callaway CS, Trevino JG, Hu H, Judge SM, Judge AR. FoxP1 is a transcriptional repressor associated with cancer cachexia that induces skeletal muscle wasting and weakness. J Cachexia Sarcopenia Muscle 2021; 12:421-442. [PMID: 33527776 PMCID: PMC8061399 DOI: 10.1002/jcsm.12666] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 12/05/2020] [Accepted: 12/16/2020] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Skeletal muscle wasting is a devastating consequence of cancer that affects up to 80% of cancer patients and associates with reduced survival. Herein, we investigated the biological significance of Forkhead box P1 (FoxP1), a transcriptional repressor that we demonstrate is up-regulated in skeletal muscle in multiple models of cancer cachexia and in cachectic cancer patients. METHODS Inducible, skeletal muscle-specific FoxP1 over-expressing (FoxP1iSkmTg/Tg ) mice were generated through crossing conditional Foxp1a transgenic mice with HSA-MCM mice that express tamoxifen-inducible Cre recombinase under control of the skeletal muscle actin promoter. To determine the requirement of FoxP1 for cancer-induced skeletal muscle wasting, FoxP1-shRNA was packaged and targeted to muscles using AAV9 delivery prior to inoculation of mice with Colon-26 Adenocarcinoma (C26) cells. RESULTS Up-regulation of FoxP1 in adult skeletal muscle was sufficient to induce features of cachexia, including 15% reduction in body mass (P < 0.05), and a 16-27% reduction in skeletal muscle mass (P < 0.05) that was characterized by a 20% reduction in muscle fibre cross-sectional area of type IIX/B muscle fibres (P = 0.020). Skeletal muscles from FoxP1iSkmTg/Tg mice also showed significant damage and myopathy characterized by the presence of centrally nucleated myofibres, extracellular matrix expansion, and were 19-26% weaker than controls (P < 0.05). Transcriptomic analysis revealed FoxP1 as a potent transcriptional repressor of skeletal muscle gene expression, with enrichment of pathways related to skeletal muscle structure and function, growth signalling, and cell quality control. Because FoxP1 functions, at least in part, as a transcriptional repressor through its interaction with histone deacetylase proteins, we treated FoxP1iSkmTg/Tg mice with Trichostatin A, and found that this completely prevented the loss of muscle mass (p = 0.007) and fibre atrophy (P < 0.001) in the tibialis anterior. In the context of cancer, FoxP1 knockdown blocked the cancer-induced repression of myocyte enhancer factor 2 (MEF2)-target genes critical to muscle differentiation and repair, improved muscle ultrastructure, and attenuated muscle fibre atrophy by 50% (P < 0.05). CONCLUSIONS In summary, we identify FoxP1 as a novel repressor of skeletal muscle gene expression that is increased in cancer cachexia, whose up-regulation is sufficient to induce skeletal muscle wasting and weakness, and required for the normal wasting response to cancer.
Collapse
Affiliation(s)
- Daria Neyroud
- Department of Physical TherapyUniversity of FloridaGainesvilleFLUSA
| | | | | | | | - Hui Hu
- Department of MicrobiologyUniversity of Alabama at BirminghamBirminghamALUSA
| | - Sarah M. Judge
- Department of Physical TherapyUniversity of FloridaGainesvilleFLUSA
| | - Andrew R. Judge
- Department of Physical TherapyUniversity of FloridaGainesvilleFLUSA
| |
Collapse
|
25
|
FOXP1 drives osteosarcoma development by repressing P21 and RB transcription downstream of P53. Oncogene 2021; 40:2785-2802. [PMID: 33716296 DOI: 10.1038/s41388-021-01742-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Revised: 02/19/2021] [Accepted: 02/26/2021] [Indexed: 01/31/2023]
Abstract
Osteosarcoma has a poor prognosis, and the poor understanding of the genetic drivers of osteosarcoma hinders further improvement in therapeutic approaches. Transcription factor forkhead box P1 (FOXP1) is a crucial modulator in skeletal development and aging. Here, we determined the role and regulatory mechanisms of FOXP1 in osteosarcoma. Higher FOXP1 expression correlated with malignancy in both osteosarcoma cell lines and clinical biopsies. FOXP1 overexpression and knockdown in osteosarcoma cell lines revealed that FOXP1 promoted proliferation, tumor sphere formation, migration and invasion, and inhibited anoikis. Mechanistically, FOXP1 acted as a repressor of P21 and RB (retinoblastoma protein) transcription, and directly interacted with the tumor suppressor p53 to inhibit its activity. Extracellular signal-regulated kinase/c-Jun N-terminal kinase (ERK/JNK) signaling and c-JUN/c-FOS transcription factors were found to be upstream activators of FOXP1. Moreover, FOXP1 silencing via lentivirus or adeno-associated virus (AAV)-mediated delivery of shRNA suppressed osteosarcoma development and progression in cell-derived and patient-derived xenograft animal models. Taken together, we demonstrate that FOXP1, which is transactivated by ERK/JNK-c-JUN/c-FOS, drives osteosarcoma development by regulating the p53-P21/RB signaling cascade, suggesting that FOXP1 is a potential target for osteosarcoma therapy.
Collapse
|
26
|
Anderson AG, Kulkarni A, Harper M, Konopka G. Single-Cell Analysis of Foxp1-Driven Mechanisms Essential for Striatal Development. Cell Rep 2021; 30:3051-3066.e7. [PMID: 32130906 PMCID: PMC7137930 DOI: 10.1016/j.celrep.2020.02.030] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 01/16/2020] [Accepted: 02/07/2020] [Indexed: 12/30/2022] Open
Abstract
The striatum is a critical forebrain structure integrating cognitive, sensory, and motor information from diverse brain regions into meaningful behavioral output. However, the transcriptional mechanisms underlying striatal development at single-cell resolution remain unknown. Using single-cell RNA sequencing (RNA-seq), we examine the cellular diversity of the early postnatal striatum and show that Foxp1, a transcription factor strongly linked to autism and intellectual disability, regulates the cellular composition, neurochemical architecture, and connectivity of the striatum in a cell-type-dependent fashion. We also identify Foxp1-regulated target genes within distinct cell types and connect these molecular changes to functional and behavioral deficits relevant to phenotypes described in patients with FOXP1 loss-of-function mutations. Using this approach, we could also examine the non-cell-autonomous effects produced by disrupting one cell type and the molecular compensation that occurs in other populations. These data reveal the cell-type-specific transcriptional mechanisms regulated by Foxp1 that underlie distinct features of striatal circuitry.
Collapse
Affiliation(s)
- Ashley G Anderson
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX 75390-9111, USA
| | - Ashwinikumar Kulkarni
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX 75390-9111, USA
| | - Matthew Harper
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX 75390-9111, USA
| | - Genevieve Konopka
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX 75390-9111, USA.
| |
Collapse
|
27
|
The Drosophila Forkhead/Fox transcription factor Jumeau mediates specific cardiac progenitor cell divisions by regulating expression of the kinesin Nebbish. Sci Rep 2021; 11:3221. [PMID: 33547352 PMCID: PMC7864957 DOI: 10.1038/s41598-021-81894-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2020] [Accepted: 12/28/2020] [Indexed: 11/16/2022] Open
Abstract
Forkhead (Fkh/Fox) domain transcription factors (TFs) mediate multiple cardiogenic processes in both mammals and Drosophila. We showed previously that the Drosophila Fox gene jumeau (jumu) controls three categories of cardiac progenitor cell division—asymmetric, symmetric, and cell division at an earlier stage—by regulating Polo kinase activity, and mediates the latter two categories in concert with the TF Myb. Those observations raised the question of whether other jumu-regulated genes also mediate all three categories of cardiac progenitor cell division or a subset thereof. By comparing microarray-based expression profiles of wild-type and jumu loss-of-function mesodermal cells, we identified nebbish (neb), a kinesin-encoding gene activated by jumu. Phenotypic analysis shows that neb is required for only two categories of jumu-regulated cardiac progenitor cell division: symmetric and cell division at an earlier stage. Synergistic genetic interactions between neb, jumu, Myb, and polo and the rescue of jumu mutations by ectopic cardiac mesoderm-specific expression of neb demonstrate that neb is an integral component of a jumu-regulated subnetwork mediating cardiac progenitor cell divisions. Our results emphasize the central role of Fox TFs in cardiogenesis and illustrate how a single TF can utilize different combinations of other regulators and downstream effectors to control distinct developmental processes.
Collapse
|
28
|
Liu XM, Du SL, Miao R, Wang LF, Zhong JC. Targeting the forkhead box protein P1 pathway as a novel therapeutic approach for cardiovascular diseases. Heart Fail Rev 2020; 27:345-355. [PMID: 32648149 DOI: 10.1007/s10741-020-09992-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Cardiovascular disease (CVD) is the leading cause of death worldwide and encompasses diverse diseases of the vasculature, myocardium, cardiac electrical circuit, and cardiac development. Forkhead box protein P1 (Foxp1) is a large multi-domain transcriptional regulator belonging to the Fox family with winged helix DNA-binding protein, which plays critical roles in cardiovascular homeostasis and disorders. The broad distribution of Foxp1 and alternative splicing isoforms implicate its distinct functions in diverse cardiac and vascular cells and tissue types. Foxp1 is essential for diverse biological processes and has been shown to regulate cellular proliferation, apoptosis, oxidative stress, fibrosis, angiogenesis, cardiovascular remodeling, and dysfunction. Notably, both loss-of-function and gain-of-function approaches have defined critical roles of Foxp1 in CVD. Genetic deletion of Foxp1 results in pathological cardiac remodeling, exacerbation of atherosclerotic lesion formation, prolonged occlusive thrombus formation, severe cardiac defects, and embryo death. In contrast, activation of Foxp1 performs a wide range of physiological effects, including cell growth, hypertrophy, differentiation, angiogenesis, and cardiac development. More importantly, Foxp1 exerts anti-inflammatory and anti-atherosclerotic effects in controlling coronary thrombus formation and myocardial infarction (MI). Thus, targeting for Foxp1 signaling has emerged as a pre-warning biomarker and a novel therapeutic approach against progression of CVD, and an increased understanding of cardiovascular actions of the Foxp1 signaling will help to develop effective interventions. In this review, we focus on the diverse actions and underlying mechanisms of Foxp1 highlighting its roles in CVD, including heart failure, MI, atherosclerosis, congenital heart defects, and atrial fibrillation.
Collapse
Affiliation(s)
- Xin-Ming Liu
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, China
| | - Sheng-Li Du
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, China
| | - Ran Miao
- Medical Research Center, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, China
| | - Le-Feng Wang
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, China.
| | - Jiu-Chang Zhong
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, China. .,Medical Research Center, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, China.
| |
Collapse
|
29
|
Machine learning uncovers cell identity regulator by histone code. Nat Commun 2020; 11:2696. [PMID: 32483223 PMCID: PMC7264183 DOI: 10.1038/s41467-020-16539-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Accepted: 05/09/2020] [Indexed: 01/13/2023] Open
Abstract
Conversion between cell types, e.g., by induced expression of master transcription factors, holds great promise for cellular therapy. Our ability to manipulate cell identity is constrained by incomplete information on cell identity genes (CIGs) and their expression regulation. Here, we develop CEFCIG, an artificial intelligent framework to uncover CIGs and further define their master regulators. On the basis of machine learning, CEFCIG reveals unique histone codes for transcriptional regulation of reported CIGs, and utilizes these codes to predict CIGs and their master regulators with high accuracy. Applying CEFCIG to 1,005 epigenetic profiles, our analysis uncovers the landscape of regulation network for identity genes in individual cell or tissue types. Together, this work provides insights into cell identity regulation, and delivers a powerful technique to facilitate regenerative medicine.
Collapse
|
30
|
Woo CC, Liu W, Lin XY, Dorajoo R, Lee KW, Richards AM, Lee CN, Wongsurawat T, Nookaew I, Sorokin V. The interaction between 30b-5p miRNA and MBNL1 mRNA is involved in vascular smooth muscle cell differentiation in patients with coronary atherosclerosis. Int J Mol Sci 2019; 21:ijms21010011. [PMID: 31861407 PMCID: PMC6982107 DOI: 10.3390/ijms21010011] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 12/12/2019] [Accepted: 12/13/2019] [Indexed: 01/20/2023] Open
Abstract
Vascular smooth muscle cells (VSMCs) in the arterial wall have diverse functions. In pathological states, the interplay between transcripts and microRNAs (miRNAs) leads to phenotypic changes. Understanding the regulatory role of miRNAs and their target genes may reveal how VSMCs modulate the pathogenesis of coronary artery disease. Laser capture microdissection was performed on aortic wall tissues obtained from coronary artery bypass graft patients with and without recent acute myocardial infarction (MI). The mSMRT-qPCR miRNA assay platform (MiRXES, Singapore) was used to profile miRNA. The miRNA data were co-analyzed with significant mRNA transcripts. TargetScan 7.1 was applied to evaluate miRNA-mRNA interactions. The miRNA profiles of 29 patients (16 MI and 13 non-MI) were evaluated. Thirteen VSMC-related miRNAs were differentially expressed between the MI and non-MI groups. Analysis revealed seven miRNA-targeted mRNAs related to muscular tissue differentiation and proliferation. TargetScan revealed that among the VSMC-related transcripts, MBNL1 had a recognition site that matched the hsa-miR-30b-5p target seed sequence. In addition to predicted analysis, our experiment in vitro with human VSMC culture confirmed that hsa-miR-30b-5p negatively correlated with MBNL1. Our data showed that overexpression of hsa-miR-30b-5p led to downregulation of MBNL1 in VSMCs. This process influences VSMC proliferation and might be involved in VSMC differentiation.
Collapse
Affiliation(s)
- Chin Cheng Woo
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore; (C.C.W.); (C.N.L.)
| | - Wenting Liu
- Genome Institute of Singapore, Agency for Science, Technology and Research (A*STAR), Singapore 138672, Singapore; (W.L.); (R.D.)
| | - Xiao Yun Lin
- Department of Cardiac, Thoracic and Vascular Surgery, National University Hospital, National University Health System, Singapore 119228, Singapore;
| | - Rajkumar Dorajoo
- Genome Institute of Singapore, Agency for Science, Technology and Research (A*STAR), Singapore 138672, Singapore; (W.L.); (R.D.)
| | - Kee Wah Lee
- Department of Pathology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore;
| | - A Mark Richards
- Cardiovascular Research Institute, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore;
- Christchurch Heart Institute, University of Otago, Christchurch 8140, New Zealand
| | - Chuen Neng Lee
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore; (C.C.W.); (C.N.L.)
- Department of Cardiac, Thoracic and Vascular Surgery, National University Hospital, National University Health System, Singapore 119228, Singapore;
| | - Thidathip Wongsurawat
- Department of Biomedical Informatics, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (T.W.); (I.N.)
| | - Intawat Nookaew
- Department of Biomedical Informatics, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (T.W.); (I.N.)
| | - Vitaly Sorokin
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore; (C.C.W.); (C.N.L.)
- Department of Cardiac, Thoracic and Vascular Surgery, National University Hospital, National University Health System, Singapore 119228, Singapore;
- Correspondence: ; Tel.: +65-6779-5555
| |
Collapse
|
31
|
Expression and prognostic value of FOXP1 in esophageal squamous cell carcinoma. Pathol Res Pract 2019; 215:152645. [DOI: 10.1016/j.prp.2019.152645] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2019] [Revised: 09/02/2019] [Accepted: 09/15/2019] [Indexed: 11/22/2022]
|
32
|
Liu P, Huang S, Ling S, Xu S, Wang F, Zhang W, Zhou R, He L, Xia X, Yao Z, Fan Y, Wang N, Hu C, Zhao X, Tucker HO, Wang J, Guo X. Foxp1 controls brown/beige adipocyte differentiation and thermogenesis through regulating β3-AR desensitization. Nat Commun 2019; 10:5070. [PMID: 31699980 PMCID: PMC6838312 DOI: 10.1038/s41467-019-12988-8] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Accepted: 10/02/2019] [Indexed: 01/08/2023] Open
Abstract
β-Adrenergic receptor (β-AR) signaling is a pathway controlling adaptive thermogenesis in brown or beige adipocytes. Here we investigate the biological roles of the transcription factor Foxp1 in brown/beige adipocyte differentiation and thermogenesis. Adipose-specific deletion of Foxp1 leads to an increase of brown adipose activity and browning program of white adipose tissues. The Foxp1-deficient mice show an augmented energy expenditure and are protected from diet-induced obesity and insulin resistance. Consistently, overexpression of Foxp1 in adipocytes impairs adaptive thermogenesis and promotes diet-induced obesity. A robust change in abundance of the β3-adrenergic receptor (β3-AR) is observed in brown/beige adipocytes from both lines of mice. Molecularly, Foxp1 directly represses β3-AR transcription and regulates its desensitization behavior. Taken together, our findings reveal Foxp1 as a master transcriptional repressor of brown/beige adipocyte differentiation and thermogenesis, and provide an important clue for its targeting and treatment of obesity. Beta3-adrenergic receptor (b3-AR) signaling in response to cold activates adipose tissue thermogenesis. Here the authors identify the transcription factor FoxP1 as a direct negative regulator of b3-AR expression and show that loss of FoxP1 leads to enhanced development of thermogenic adipose tissue.
Collapse
Affiliation(s)
- Pei Liu
- Department of Nephrology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Sixia Huang
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Shifeng Ling
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Shuqin Xu
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Fuhua Wang
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Wei Zhang
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Rujiang Zhou
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Lin He
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Xuechun Xia
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Zhengju Yao
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Ying Fan
- Department of Nephrology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Niansong Wang
- Department of Nephrology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Congxia Hu
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Xiaodong Zhao
- Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Haley O Tucker
- Institute for Cellular and Molecular Biology, University of Texas at Austin, Austin, TX, 78712, USA
| | - Jiqiu Wang
- Department of Endocrinology and Metabolism, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Xizhi Guo
- Department of Nephrology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China. .,Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, 200240, China.
| |
Collapse
|
33
|
Affiliation(s)
- Jenny E Kanter
- Department of Medicine, Division of Metabolism, Endocrinology and Nutrition, UW Medicine Diabetes Institute, University of Washington School of Medicine, Seattle, WA
| |
Collapse
|
34
|
Zhuang T, Liu J, Chen X, Zhang L, Pi J, Sun H, Li L, Bauer R, Wang H, Yu Z, Zhang Q, Tomlinson B, Chan P, Zheng X, Morrisey E, Liu Z, Reilly M, Zhang Y. Endothelial Foxp1 Suppresses Atherosclerosis via Modulation of Nlrp3 Inflammasome Activation. Circ Res 2019; 125:590-605. [PMID: 31318658 DOI: 10.1161/circresaha.118.314402] [Citation(s) in RCA: 110] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
RATIONALE Endothelial dysfunction results in sustained and chronic vascular inflammation, which is central to atherosclerotic diseases. However, transcriptional regulation of vascular endothelial inflammation has not been well clarified. OBJECTIVE This study aims to explore Foxp (forkhead box P) transcription factor 1 in regulation of endothelial homeostasis, atherogenesis, and its mechanisms. METHODS AND RESULTS To assess the importance of Foxp1 in atherosclerosis, Foxp1 expression was analyzed in human coronary artery and mouse artery, and we observed significant downregulation of Foxp1 in atherosclerotic and atherosusceptible endothelium. Endothelial-specific Foxp1 knockout mice (Foxp1ECKO) were bred onto ApoeKO mice to generate endothelial Foxp1-deletion hyperlipidemic model Foxp1ECKO;ApoeKO, which displayed significant increases in atherosclerotic lesion formation in aortas and aortic roots with enhanced monocyte adhesion, migration, and infiltration into the vascular wall and formation of inflammatory lipid-laden macrophages. In contrast, endothelial-specific Foxp1 overexpression mice Foxp1ECTg;ApoeKO exhibited reduced atherosclerotic lesion formation with less monocyte infiltration. Foxp1 was further identified as a gatekeeper of vessel inflammation by direct regulation of endothelial inflammasome components, including Nlrp3 (NLR [nucleotide-binding and leucine-rich repeat immune receptors] family pyrin domain containing 3), caspase-1, and IL (interleukin)-1β. Moreover, endothelial Foxp1 was found to be regulated by Klf2 (Kruppel-like factor 2). Oscillatory shear stress downregulated Foxp1 expression via repressing Klf2 expression in endothelium, and, therefore, promoted endothelial inflammasome activation, leading to atherosclerotic lesion formation. Simvastatin upregulated the reduced expression of Klf2 and Foxp1 in atherosusceptible vascular endothelium and alleviated vascular inflammation contributing to its inhibitory effect in atherosclerosis. CONCLUSIONS These data are the first in vivo experimental validation of an atheroprotective role of endothelial Klf2 and Foxp1, which reveals a Klf2-Foxp1 transcriptional network in endothelial cells as a novel regulator of endothelial inflammasome activation for atherogenesis, therefore, provides opportunities for therapeutic intervention of atherosclerotic diseases and uncovers a novel atheroprotective mechanism for simvastatin.
Collapse
Affiliation(s)
- Tao Zhuang
- From the Key Laboratory of Arrhythmias of the Ministry of Education of China, Research Center for Translational Medicine (T.Z., J.L., X.C., L.Z., H.S., Z.Y., Z.L., Y.Z.), Shanghai East Hospital, Tongji University School of Medicine, China
| | - Jie Liu
- From the Key Laboratory of Arrhythmias of the Ministry of Education of China, Research Center for Translational Medicine (T.Z., J.L., X.C., L.Z., H.S., Z.Y., Z.L., Y.Z.), Shanghai East Hospital, Tongji University School of Medicine, China
| | - Xiaoli Chen
- From the Key Laboratory of Arrhythmias of the Ministry of Education of China, Research Center for Translational Medicine (T.Z., J.L., X.C., L.Z., H.S., Z.Y., Z.L., Y.Z.), Shanghai East Hospital, Tongji University School of Medicine, China
| | - Lin Zhang
- From the Key Laboratory of Arrhythmias of the Ministry of Education of China, Research Center for Translational Medicine (T.Z., J.L., X.C., L.Z., H.S., Z.Y., Z.L., Y.Z.), Shanghai East Hospital, Tongji University School of Medicine, China
| | - Jingjiang Pi
- Cardiology (J.P., Q.Z.), Shanghai East Hospital, Tongji University School of Medicine, China
| | - Huimin Sun
- From the Key Laboratory of Arrhythmias of the Ministry of Education of China, Research Center for Translational Medicine (T.Z., J.L., X.C., L.Z., H.S., Z.Y., Z.L., Y.Z.), Shanghai East Hospital, Tongji University School of Medicine, China
| | - Li Li
- Cardiovascular Institute, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia (L.L.)
| | - Robert Bauer
- Cardiology Division, Department of Medicine (R.B.), Columbia University, New York, NY
| | - Haikun Wang
- Key Laboratory of Molecular Virology and Immunology, Institute Pasteur of Shanghai, Chinese Academy of Sciences, University of Chinese Academy of Sciences, China (H.W.)
| | - Zuoren Yu
- From the Key Laboratory of Arrhythmias of the Ministry of Education of China, Research Center for Translational Medicine (T.Z., J.L., X.C., L.Z., H.S., Z.Y., Z.L., Y.Z.), Shanghai East Hospital, Tongji University School of Medicine, China
| | - Qi Zhang
- Cardiology (J.P., Q.Z.), Shanghai East Hospital, Tongji University School of Medicine, China
| | - Brian Tomlinson
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong SAR (B.T.)
| | - Paul Chan
- Division of Cardiology, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taiwan (P.C.)
| | - Xiangjian Zheng
- Department of Pharmacology, School of Basic Medical Sciences, Tianjin Medical University (X.Z.).,Laboratory of Cardiovascular Signaling, Centenary Institute, and Sydney Medical School, University of Sydney, Sydney, NSW, Australia (X.Z.)
| | - Edward Morrisey
- Department of Cell and Developmental Biology (E.M.), University of Pennsylvania, Philadelphia.,Department of Medicine (E.M.), University of Pennsylvania, Philadelphia.,Penn Cardiovascular Institute (E.M.), University of Pennsylvania, Philadelphia.,Penn Institute for Regenerative Medicine (E.M.), University of Pennsylvania, Philadelphia
| | - Zhongmin Liu
- From the Key Laboratory of Arrhythmias of the Ministry of Education of China, Research Center for Translational Medicine (T.Z., J.L., X.C., L.Z., H.S., Z.Y., Z.L., Y.Z.), Shanghai East Hospital, Tongji University School of Medicine, China
| | - Muredach Reilly
- Cardiology Division, Department of Medicine and the Irving Institute for Clinical and Translational Research (M.R.), Columbia University, New York, NY
| | - Yuzhen Zhang
- From the Key Laboratory of Arrhythmias of the Ministry of Education of China, Research Center for Translational Medicine (T.Z., J.L., X.C., L.Z., H.S., Z.Y., Z.L., Y.Z.), Shanghai East Hospital, Tongji University School of Medicine, China
| |
Collapse
|
35
|
Doyle MJ, Magli A, Estharabadi N, Amundsen D, Mills LJ, Martin CM. Sox7 Regulates Lineage Decisions in Cardiovascular Progenitor Cells. Stem Cells Dev 2019; 28:1089-1103. [PMID: 31154937 DOI: 10.1089/scd.2019.0040] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Specification of the mesodermal lineages requires a complex set of morphogenetic events orchestrated by interconnected signaling pathways and gene regulatory networks. The transcription factor Sox7 has critical functions in differentiation of multiple mesodermal lineages, including cardiac, endothelial, and hematopoietic. Using a doxycycline-inducible mouse embryonic stem cell line, we have previously shown that expression of Sox7 in cardiovascular progenitor cells promotes expansion of endothelial progenitor cells (EPCs). In this study, we show that the ability of Sox7 to promote endothelial cell fate occurs at the expense of the cardiac lineage. Using ChIP-Seq coupled with ATAC-Seq we identify downstream target genes of Sox7 in cardiovascular progenitor cells and by integrating these data with transcriptomic analyses, we define Sox7-dependent gene programs specific to cardiac and EPCs. Furthermore, we demonstrate a protein-protein interaction between SOX7 and GATA4 and provide evidence that SOX7 interferes with the transcriptional activity of GATA4 on cardiac genes. In addition, we show that Sox7 modulates WNT and BMP signaling during cardiovascular differentiation. Our data represent the first genome-wide analysis of Sox7 function and reveal a critical role for Sox7 in regulating signaling pathways that affect cardiovascular progenitor cell differentiation.
Collapse
Affiliation(s)
- Michelle J Doyle
- 1Department of Medicine, University of Minnesota, Minneapolis, Minnesota.,2Lillehei Heart Institute, University of Minnesota, Minneapolis, Minnesota
| | - Alessandro Magli
- 2Lillehei Heart Institute, University of Minnesota, Minneapolis, Minnesota.,3Stem Cell Institute, University of Minnesota, Minneapolis, Minnesota
| | - Nima Estharabadi
- 1Department of Medicine, University of Minnesota, Minneapolis, Minnesota.,2Lillehei Heart Institute, University of Minnesota, Minneapolis, Minnesota
| | - Danielle Amundsen
- 1Department of Medicine, University of Minnesota, Minneapolis, Minnesota.,2Lillehei Heart Institute, University of Minnesota, Minneapolis, Minnesota
| | - Lauren J Mills
- 4Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota
| | - Cindy M Martin
- 1Department of Medicine, University of Minnesota, Minneapolis, Minnesota.,2Lillehei Heart Institute, University of Minnesota, Minneapolis, Minnesota
| |
Collapse
|
36
|
Liu J, Zhuang T, Pi J, Chen X, Zhang Q, Li Y, Wang H, Shen Y, Tomlinson B, Chan P, Yu Z, Cheng Y, Zheng X, Reilly M, Morrisey E, Zhang L, Liu Z, Zhang Y. Endothelial Forkhead Box Transcription Factor P1 Regulates Pathological Cardiac Remodeling Through Transforming Growth Factor-β1-Endothelin-1 Signal Pathway. Circulation 2019; 140:665-680. [PMID: 31177814 DOI: 10.1161/circulationaha.119.039767] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Pathological cardiac fibrosis and hypertrophy, the common features of left ventricular remodeling, often progress to heart failure. Forkhead box transcription factor P1 (Foxp1) in endothelial cells (ECs) has been shown to play an important role in heart development. However, the effect of EC-Foxp1 on pathological cardiac remodeling has not been well clarified. This study aims to determine the role of EC-Foxp1 in pathological cardiac remodeling and the underlying mechanisms. METHODS Foxp1 EC-specific loss-of-function and gain-of-function mice were generated, and an angiotensin II infusion or a transverse aortic constriction operation mouse model was used to study the cardiac remodeling mechanisms. Foxp1 downstream target gene transforming growth factor-β1 (TGF-β1) was confirmed by chromatin immunoprecipitation and luciferase assays. Finally, the effects of TGF-β1 blockade on EC-Foxp1 deletion-mediated profibrotic and prohypertrophic phenotypic changes were further confirmed by pharmacological inhibition, more specifically by RGD-peptide magnetic nanoparticle target delivery of TGF-β1-siRNA to ECs. RESULTS Foxp1 expression is significantly downregulated in cardiac ECs during angiotensin II-induced cardiac remodeling. EC-Foxp1 deletion results in severe cardiac remodeling, including more cardiac fibrosis with myofibroblast formation and extracellular matrix protein production, as well as decompensated cardiac hypertrophy and further exacerbation of cardiac dysfunction on angiotensin II infusion or transverse aortic constriction operation. In contrast, EC-Foxp1 gain of function protects against pathological cardiac remodeling and improves cardiac dysfunction. TGF-β1 signals are identified as Foxp1 direct target genes, and EC-Foxp1 deletion upregulates TGF-β1 signals to promote myofibroblast formation through fibroblast proliferation and transformation, resulting in severe cardiac fibrosis. Moreover, EC-Foxp1 deletion enhances TGF-β1-promoted endothelin-1 expression, which significantly increases cardiomyocyte size and reactivates cardiac fetal genes, leading to pathological cardiac hypertrophy. Correspondingly, these EC-Foxp1 deletion-mediated profibrotic and prohypertrophic phenotypic changes and cardiac dysfunction are normalized by the blockade of TGF-β1 signals through pharmacological inhibition and RGD-peptide magnetic nanoparticle target delivery of TGF-β1-siRNA to ECs. CONCLUSIONS EC-Foxp1 regulates the TGF-β1-endothelin-1 pathway to control pathological cardiac fibrosis and hypertrophy, resulting in cardiac dysfunction. Therefore, targeting the EC-Foxp1-TGF-β1-endothelin-1 pathway might provide a future novel therapy for heart failure.
Collapse
Affiliation(s)
- Jie Liu
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Research Center for Translational Medicine (J.L., T.Z., X.C., Z.Y., L.Z., Z.L., Y.Z.), Tongji University School of Medicine, China
| | - Tao Zhuang
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Research Center for Translational Medicine (J.L., T.Z., X.C., Z.Y., L.Z., Z.L., Y.Z.), Tongji University School of Medicine, China
| | - Jingjiang Pi
- Department of Cardiology (J.P., Q.Z., Y.L.), Shanghai East Hospital, Tongji University School of Medicine, China
| | - Xiaoli Chen
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Research Center for Translational Medicine (J.L., T.Z., X.C., Z.Y., L.Z., Z.L., Y.Z.), Tongji University School of Medicine, China
| | - Qi Zhang
- Department of Cardiology (J.P., Q.Z., Y.L.), Shanghai East Hospital, Tongji University School of Medicine, China
| | - Ying Li
- Department of Cardiology (J.P., Q.Z., Y.L.), Shanghai East Hospital, Tongji University School of Medicine, China
| | - Haikun Wang
- CAS Key Laboratory of Molecular Virology and Immunology, Institute Pasteur of Shanghai, Chinese Academy of Science, University of Chinese Academy of Sciences (H.W.)
| | - Yajing Shen
- Institute for Biomedical Engineering and Nano Science (Y.S., Y.C.), Tongji University School of Medicine, China
| | - Brian Tomlinson
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong (B.T.)
| | - Paul Chan
- Division of Cardiology, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taiwan (P.C.)
| | - Zuoren Yu
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Research Center for Translational Medicine (J.L., T.Z., X.C., Z.Y., L.Z., Z.L., Y.Z.), Tongji University School of Medicine, China
| | - Yu Cheng
- Institute for Biomedical Engineering and Nano Science (Y.S., Y.C.), Tongji University School of Medicine, China
| | - Xiangjian Zheng
- Department of Pharmacology, School of Basic Medical Sciences, Tianjin Medical University, China (X.Z.).,Laboratory of Cardiovascular Signaling, Centenary Institute, Camperdown, Australia (X.Z.)
| | - Muredach Reilly
- Cardiology Division, Department of Medicine and the Irving Institute for Clinical and Translational Research, Columbia University, New York (M.R.)
| | - Edward Morrisey
- Department of Cell and Developmental Biology, Department of Medicine, Penn Cardiovascular Institute, and Penn Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia (E.M.)
| | - Lin Zhang
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Research Center for Translational Medicine (J.L., T.Z., X.C., Z.Y., L.Z., Z.L., Y.Z.), Tongji University School of Medicine, China
| | - Zhongmin Liu
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Research Center for Translational Medicine (J.L., T.Z., X.C., Z.Y., L.Z., Z.L., Y.Z.), Tongji University School of Medicine, China
| | - Yuzhen Zhang
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Research Center for Translational Medicine (J.L., T.Z., X.C., Z.Y., L.Z., Z.L., Y.Z.), Tongji University School of Medicine, China
| |
Collapse
|
37
|
Payan SM, Hubert F, Rochais F. Cardiomyocyte proliferation, a target for cardiac regeneration. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2019; 1867:118461. [PMID: 30930108 DOI: 10.1016/j.bbamcr.2019.03.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 02/20/2019] [Accepted: 03/13/2019] [Indexed: 12/23/2022]
Abstract
Cardiac diseases, characterized by cardiomyocyte loss, lead to dramatic impairment of cardiac function and ultimately to congestive heart failure. Despite significant advances, conventional treatments do not correct the defects in cardiac muscle cell numbers and the prognosis of congestive heart failure remains poor. The existence, in adult mammalian heart, of low but detectable cardiomyocyte proliferative capacities has shifted the target of regenerative therapy toward new therapeutical strategy. Indeed, the stimulation of terminally differentiated cardiomyocyte proliferation represents the main therapeutic approach for heart regeneration. Increasing evidence demonstrating that the loss of mammalian cardiomyocyte renewal potential shortly after birth causes the loss of regenerative capacities, strongly support the hypothesis that a detailed understanding of the molecular mechanisms controlling fetal and postnatal cardiomyocyte proliferation is essential to identify targets for cardiac regeneration. Here, we will review major developmental mechanisms regulating fetal cardiomyocyte proliferation and will describe the impact of the developmental switch, operating at birth and driving postnatal heart maturation, on the regulation of adult cardiomyocyte proliferation, all these mechanisms representing potential targets for cardiac repair and regeneration.
Collapse
Affiliation(s)
- Sandy M Payan
- Aix-Marseille Univ, INSERM, MMG, U 1251, Marseille, France
| | - Fabien Hubert
- Aix-Marseille Univ, INSERM, MMG, U 1251, Marseille, France
| | | |
Collapse
|
38
|
Konopacki C, Pritykin Y, Rubtsov Y, Leslie CS, Rudensky AY. Transcription factor Foxp1 regulates Foxp3 chromatin binding and coordinates regulatory T cell function. Nat Immunol 2019; 20:232-242. [PMID: 30643266 DOI: 10.1038/s41590-018-0291-z] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Accepted: 11/25/2018] [Indexed: 12/22/2022]
Abstract
Regulatory T cells (Treg cells), whose differentiation and function are controlled by transcription factor Foxp3, express the closely related family member Foxp1. Here we explored Foxp1 function in Treg cells. We found that a large number of Foxp3-bound genomic sites in Treg cells were occupied by Foxp1 in both Treg cells and conventional T cells (Tconv cells). In Treg cells, Foxp1 markedly increased Foxp3 binding to these sites. Foxp1 deficiency in Treg cells resulted in their impaired function and competitive fitness, associated with markedly reduced CD25 expression and interleukin-2 (IL-2) responsiveness, diminished CTLA-4 expression and increased SATB1 expression. The characteristic expression patterns of CD25, Foxp3 and CTLA-4 in Treg cells were fully or partially rescued by strong IL-2 signaling. Our studies suggest that Foxp1 serves an essential non-redundant function in Treg cells by enforcing Foxp3-mediated regulation of gene expression and enabling efficient IL-2 signaling in these cells.
Collapse
Affiliation(s)
- Catherine Konopacki
- Howard Hughes Medical Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA.,Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Yuri Pritykin
- Computational and Systems Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Yury Rubtsov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia.,Faculty of Biology and Biotechnology, National Research University Higher School of Economics, Moscow, Russia
| | - Christina S Leslie
- Computational and Systems Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| | - Alexander Y Rudensky
- Howard Hughes Medical Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA. .,Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA. .,Ludwig Center at Memorial Sloan Kettering Cancer Center, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
39
|
Fu NY, Pal B, Chen Y, Jackling FC, Milevskiy M, Vaillant F, Capaldo BD, Guo F, Liu KH, Rios AC, Lim N, Kueh AJ, Virshup DM, Herold MJ, Tucker HO, Smyth GK, Lindeman GJ, Visvader JE. Foxp1 Is Indispensable for Ductal Morphogenesis and Controls the Exit of Mammary Stem Cells from Quiescence. Dev Cell 2018; 47:629-644.e8. [PMID: 30523786 DOI: 10.1016/j.devcel.2018.10.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Revised: 08/28/2018] [Accepted: 10/01/2018] [Indexed: 12/12/2022]
Abstract
Long-lived quiescent mammary stem cells (MaSCs) are presumed to coordinate the dramatic expansion of ductal epithelium that occurs through the different phases of postnatal development, but little is known about the molecular regulators that underpin their activation. We show that ablation of the transcription factor Foxp1 in the mammary gland profoundly impairs ductal morphogenesis, resulting in a rudimentary tree throughout life. Foxp1-deficient glands were highly enriched for quiescent Tspan8hi MaSCs, which failed to become activated even in competitive transplantation assays, thus highlighting a cell-intrinsic defect. Foxp1 deletion also resulted in aberrant expression of basal genes in luminal cells, inferring a role in cell-fate decisions. Notably, Foxp1 was uncovered as a direct repressor of Tspan8 in basal cells, and deletion of Tspan8 rescued the defects in ductal morphogenesis elicited by Foxp1 loss. Thus, a single transcriptional regulator Foxp1 can control the exit of MaSCs from dormancy to orchestrate differentiation and development.
Collapse
Affiliation(s)
- Nai Yang Fu
- Stem Cells and Cancer Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, The University of Melbourne, Parkville, VIC 3010, Australia; Cancer and Stem Cell Biology Program, Duke-NUS Medical School, Singapore 169857, Singapore.
| | - Bhupinder Pal
- Stem Cells and Cancer Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Yunshun Chen
- Department of Medical Biology, The University of Melbourne, Parkville, VIC 3010, Australia; Bioinformatics Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
| | - Felicity C Jackling
- Stem Cells and Cancer Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Michael Milevskiy
- Stem Cells and Cancer Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - François Vaillant
- Stem Cells and Cancer Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Bianca D Capaldo
- Stem Cells and Cancer Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Fusheng Guo
- Cancer and Stem Cell Biology Program, Duke-NUS Medical School, Singapore 169857, Singapore
| | - Kevin H Liu
- Stem Cells and Cancer Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Anne C Rios
- Stem Cells and Cancer Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, The University of Melbourne, Parkville, VIC 3010, Australia; Prinses Máxima Center for Pediatric Oncology, Hubrecht Institute, Uppsalalaan 8, Utrecht 3584 CT, the Netherlands
| | - Nicholas Lim
- Cancer and Stem Cell Biology Program, Duke-NUS Medical School, Singapore 169857, Singapore
| | - Andrew J Kueh
- Department of Medical Biology, The University of Melbourne, Parkville, VIC 3010, Australia; Division of Molecular Genetics of Cancer Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
| | - David M Virshup
- Cancer and Stem Cell Biology Program, Duke-NUS Medical School, Singapore 169857, Singapore
| | - Marco J Herold
- Department of Medical Biology, The University of Melbourne, Parkville, VIC 3010, Australia; Division of Molecular Genetics of Cancer Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
| | - Haley O Tucker
- Molecular Biosciences and Institute for Molecular and Cellular Biology, University of Texas at Austin, Austin, TX, USA
| | - Gordon K Smyth
- Bioinformatics Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; School of Mathematics and Statistics, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Geoffrey J Lindeman
- Stem Cells and Cancer Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; The Royal Melbourne Hospital and Peter MacCallum Cancer Centre, Parkville, VIC 3050, Australia; Department of Medicine, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Jane E Visvader
- Stem Cells and Cancer Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, The University of Melbourne, Parkville, VIC 3010, Australia.
| |
Collapse
|
40
|
Abstract
Ventricular myocardial development is a well-orchestrated process involving different cardiac structures, multiple signal pathways, and myriad proteins. Dysregulation of this important developmental event can result in cardiomyopathies, such as left ventricle non-compaction, which affect the pediatric population and the adults. Human and mouse studies have shed light upon the etiology of some cardiomyopathy cases and highlighted the contribution of both genetic and environmental factors. However, the regulation of ventricular myocardial development remains incompletely understood. Zinc is an essential trace metal with structural, enzymatic, and signaling function. Perturbation of zinc homeostasis has resulted in developmental and physiological defects including cardiomyopathy. In this review, we summarize several mechanisms by which zinc and zinc transporters can impact the regulation of ventricular myocardial development. Based on our review, we propose that zinc deficiency and mutations of zinc transporters may underlie some cardiomyopathy cases especially those involving ventricular myocardial development defects.
Collapse
Affiliation(s)
- Wen Lin
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY, 10591, USA
| | - Deqiang Li
- Division of Cardiac Surgery, School of Medicine, University of Maryland, 800 West Baltimore ST, Rm 314, Baltimore, MD, 21201, USA.
| |
Collapse
|
41
|
Kim KH, Nakaoka Y, Augustin HG, Koh GY. Myocardial Angiopoietin-1 Controls Atrial Chamber Morphogenesis by Spatiotemporal Degradation of Cardiac Jelly. Cell Rep 2018; 23:2455-2466. [DOI: 10.1016/j.celrep.2018.04.080] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Revised: 03/09/2018] [Accepted: 04/17/2018] [Indexed: 02/02/2023] Open
|
42
|
Bond J, Domaschenz R, Roman-Trufero M, Sabbattini P, Ferreiros-Vidal I, Gerrard G, Asnafi V, Macintyre E, Merkenschlager M, Dillon N. Direct interaction of Ikaros and Foxp1 modulates expression of the G protein-coupled receptor G2A in B-lymphocytes and acute lymphoblastic leukemia. Oncotarget 2018; 7:65923-65936. [PMID: 27588474 PMCID: PMC5323203 DOI: 10.18632/oncotarget.11688] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Accepted: 08/13/2016] [Indexed: 12/13/2022] Open
Abstract
Ikaros and Foxp1 are transcription factors that play key roles in normal lymphopoiesis and lymphoid malignancies. We describe a novel physical and functional interaction between the proteins, which requires the central zinc finger domain of Ikaros. The Ikaros-Foxp1 interaction is abolished by deletion of this region, which corresponds to the IK6 isoform that is commonly associated with high-risk acute lymphoblastic leukemia (ALL). We also identify the Gpr132 gene, which encodes the orphan G protein-coupled receptor G2A, as a novel target for Foxp1. Increased expression of Foxp1 enhanced Gpr132 transcription and caused cell cycle changes, including G2 arrest. Co-expression of wild-type Ikaros, but not IK6, displaced Foxp1 binding from the Gpr132 gene, reversed the increase in Gpr132 expression and inhibited G2 arrest. Analysis of primary ALL samples revealed a significant increase in GPR132 expression in IKZF1-deleted BCR-ABL negative patients, suggesting that levels of wild-type Ikaros may influence the regulation of G2A in B-ALL. Our results reveal a novel effect of Ikaros haploinsufficiency on Foxp1 functioning, and identify G2A as a potential modulator of the cell cycle in Ikaros-deleted B-ALL.
Collapse
Affiliation(s)
- Jonathan Bond
- Gene Regulation and Chromatin Group, MRC Clinical Sciences Centre, Imperial College Faculty of Medicine, Hammersmith Campus, London W12 0NN, United Kingdom.,Université Paris Descartes Sorbonne Cité, Institut Necker-Enfants Malades (INEM), Institut National de Recherche Médicale (INSERM), and Laboratory of Onco-Hematology, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Necker Enfants-Malades, Paris, France
| | - Renae Domaschenz
- Gene Regulation and Chromatin Group, MRC Clinical Sciences Centre, Imperial College Faculty of Medicine, Hammersmith Campus, London W12 0NN, United Kingdom.,Present address: Chromatin and Transcriptional Regulation Group, John Curtin School of Medical Research, The Australian National University, Canberra, Australian Capital Territory, Australia
| | - Mónica Roman-Trufero
- Gene Regulation and Chromatin Group, MRC Clinical Sciences Centre, Imperial College Faculty of Medicine, Hammersmith Campus, London W12 0NN, United Kingdom
| | - Pierangela Sabbattini
- Gene Regulation and Chromatin Group, MRC Clinical Sciences Centre, Imperial College Faculty of Medicine, Hammersmith Campus, London W12 0NN, United Kingdom
| | - Isabel Ferreiros-Vidal
- Lymphocyte Development Group, MRC Clinical Sciences Centre, Imperial College Faculty of Medicine, Hammersmith Campus, London W12 0NN, United Kingdom
| | - Gareth Gerrard
- Imperial Molecular Pathology, Imperial College Academic Health Sciences Centre, Hammersmith Campus, London W12 0NN, United Kingdom
| | - Vahid Asnafi
- Université Paris Descartes Sorbonne Cité, Institut Necker-Enfants Malades (INEM), Institut National de Recherche Médicale (INSERM), and Laboratory of Onco-Hematology, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Necker Enfants-Malades, Paris, France
| | - Elizabeth Macintyre
- Université Paris Descartes Sorbonne Cité, Institut Necker-Enfants Malades (INEM), Institut National de Recherche Médicale (INSERM), and Laboratory of Onco-Hematology, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Necker Enfants-Malades, Paris, France
| | - Matthias Merkenschlager
- Lymphocyte Development Group, MRC Clinical Sciences Centre, Imperial College Faculty of Medicine, Hammersmith Campus, London W12 0NN, United Kingdom
| | - Niall Dillon
- Gene Regulation and Chromatin Group, MRC Clinical Sciences Centre, Imperial College Faculty of Medicine, Hammersmith Campus, London W12 0NN, United Kingdom
| |
Collapse
|
43
|
Wang S, Li Y, Jiang C, Tian H. Fibroblast growth factor 9 subfamily and the heart. Appl Microbiol Biotechnol 2017; 102:605-613. [PMID: 29198068 DOI: 10.1007/s00253-017-8652-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Revised: 11/13/2017] [Accepted: 11/13/2017] [Indexed: 12/22/2022]
Abstract
The fibroblast growth factor (FGF) 9 subfamily is a member of the FGF family, including FGF9, 16, and 20, potentially sharing similar biochemical functions due to their high degree of sequence homology. Unlike other secreted proteins which have a cleavable N-terminal secreted signal peptide, FGF9/16/20 have non-cleaved N-terminal signal peptides. As an intercellular signaling molecule, they are involved in a variety of complex responses in animal development. Cardiogenesis is controlled by many members of the transcription factor family. Evidence suggests that FGF signaling, including the FGF9 subfamily, has a pretty close association with these cardiac-specific genes. In addition, recent studies have shown that the FGF9 subfamily maintains functional adaptation and survival after myocardial infarction in adult myocardium. Since FGF9/16/20 are secreted proteins, their function characterization in cardiac regeneration can promote their potential to be developed for the treatment of cardioprotection and revascularization. Here, we conclude that the FGF9 subfamily roles in cardiac development and maintenance of postnatal cardiac homeostasis, especially cardiac function maturation and functional maintenance of the heart after injury.
Collapse
Affiliation(s)
- Shen Wang
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Yong Li
- Department of Biochemistry, Institute of Integrative Biology, University of Liverpool, Liverpool, L69 7ZB, UK
| | - Chao Jiang
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China. .,Biomedicine Collaborative Innovation Center, Wenzhou University, Wenzhou, Zhejiang, 325035, China.
| | - Haishan Tian
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China.
| |
Collapse
|
44
|
Myers A, du Souich C, Yang CL, Borovik L, Mwenifumbo J, Rupps R, Study C, Lehman A, Boerkoel CF. FOXP1 haploinsufficiency: Phenotypes beyond behavior and intellectual disability? Am J Med Genet A 2017; 173:3172-3181. [PMID: 28884888 DOI: 10.1002/ajmg.a.38462] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Revised: 05/02/2017] [Accepted: 08/04/2017] [Indexed: 01/23/2023]
Abstract
The forkhead box (FOX) transcription factors have roles in development, carcinogenesis, metabolism, and immunity. In humans FOXP1 mutations have been associated with language and speech defects, intellectual disability, autism spectrum disorder, facial dysmorphisms, and congenital anomalies of the kidney and urinary tract. In mice, Foxp1 plays critical roles in development of the spinal motor neurons, lymphocytes, cardiomyocytes, foregut, and skeleton. We hypothesized therefore that mutations of FOXP1 affect additional tissues in some humans. Supporting this hypothesis, we describe two individuals with novel variants of FOXP1 (NM_032682.5:c.975-2A>C and NM_032682.5:c.1574G>A) and additional features. One had a lung disease resembling neuroendocrine cell hyperplasia of infancy (NEHI), and the second had a skeletal disorder with undertubulation of the long bones and relapsing-remitting fevers associated with flushing and edema. Although attribution of these traits to mutation of FOXP1 requires ascertainment of additional patients, we hypothesize that the variable expression of these additional features might arise by means of stochastic developmental variation.
Collapse
Affiliation(s)
- Angela Myers
- Department of Pediatrics, University of South Dakota and Sanford Health, Sioux Falls, South Dakota
| | - Christèle du Souich
- Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia, Canada
- BC Children's Hospital Research Institute, Children's and Women's Health Centre of British Columbia, Vancouver, British Columbia, Canada
| | - Connie L Yang
- Division of Respiratory Medicine, Department of Pediatrics, University of British Columbia, and Children's and Women's Health Centre of British Columbia, Vancouver, British Columbia, Canada
| | - Lior Borovik
- Department of Genetic Counseling, Sanford Health, Sioux Falls, South Dakota
| | - Jill Mwenifumbo
- BC Children's Hospital Research Institute, Children's and Women's Health Centre of British Columbia, Vancouver, British Columbia, Canada
| | - Rosemarie Rupps
- Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia, Canada
- BC Children's Hospital Research Institute, Children's and Women's Health Centre of British Columbia, Vancouver, British Columbia, Canada
| | - Causes Study
- BC Children's Hospital Research Institute, Children's and Women's Health Centre of British Columbia, Vancouver, British Columbia, Canada
| | - Anna Lehman
- Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia, Canada
- BC Children's Hospital Research Institute, Children's and Women's Health Centre of British Columbia, Vancouver, British Columbia, Canada
| | - Cornelius F Boerkoel
- Department of Pediatrics, University of South Dakota and Sanford Health, Sioux Falls, South Dakota
- Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia, Canada
- BC Children's Hospital Research Institute, Children's and Women's Health Centre of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
45
|
Xing T, Du L, Zhuang X, Zhang L, Hao J, Wang J. Upregulation of microRNA-206 induces apoptosis of vascular smooth muscle cells and decreases risk of atherosclerosis through modulating FOXP1. Exp Ther Med 2017; 14:4097-4103. [PMID: 29104627 PMCID: PMC5658685 DOI: 10.3892/etm.2017.5071] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Accepted: 12/09/2016] [Indexed: 01/14/2023] Open
Abstract
Forkhead box protein subfamily P (FOXP) 1 has an important role in the control of gene transcription and is also reported to function as a tumor suppressor. The aim of the present study was to explore the regulatory mechanisms of atherosclerosis by investigating the function of microRNA-206 (miR-206) and the regulatory association between miR-206 and its potential target gene, FOXP1, in vascular smooth muscle cells (VSMCs). Bioinformatics tools were utilized to identify FOXP1 as a target of miR-206. Luciferase reporter analysis was used to confirm this relationship and to identify the miR-206 binding site in the FOXP1 3′-untranslated region. It was demonstrated that the relative survival rate of VSMCs was suppressed by miR-206 compared with scramble controls. Furthermore, reduced expression of miR-206 in atherosclerosis tissue samples was observed, and the mRNA and protein expression levels of FOXP1 were upregulated in atherosclerosis tissue samples, both compared with the controls, indicating a negative correlation between miR-206 and FOXP1. Additionally, when treated with miR-206 mimics, the relative survival rate of VSMCs was notably reduced, which was rescued by overexpression of FOXP1. These findings increased the understanding of the regulatory role of miR-206 in atherosclerosis in VSMCs via targeting the FOXP1 gene; therefore, intervention with miR-206 as a therapeutic technique may be a strategy for atherosclerosis treatment in the future.
Collapse
Affiliation(s)
- Tao Xing
- Department of Neurosurgery, Liaocheng People's Hospital, Liaocheng, Shandong 252000, P.R. China
| | - Lixin Du
- Department of Neurology, Liaocheng People's Hospital, Liaocheng, Shandong 252000, P.R. China
| | - Xianbo Zhuang
- Department of Neurology, Liaocheng People's Hospital, Liaocheng, Shandong 252000, P.R. China
| | - Liyong Zhang
- Department of Neurosurgery, Liaocheng People's Hospital, Liaocheng, Shandong 252000, P.R. China
| | - Jiheng Hao
- Department of Neurosurgery, Brain Hospital of Liaocheng, Liaocheng, Shandong 252000, P.R. China
| | - Jiyue Wang
- Department of Neurosurgery, Brain Hospital of Liaocheng, Liaocheng, Shandong 252000, P.R. China
| |
Collapse
|
46
|
D'Souza A, Pearman CM, Wang Y, Nakao S, Logantha SJRJ, Cox C, Bennett H, Zhang Y, Johnsen AB, Linscheid N, Poulsen PC, Elliott J, Coulson J, McPhee J, Robertson A, da Costa Martins PA, Kitmitto A, Wisløff U, Cartwright EJ, Monfredi O, Lundby A, Dobrzynski H, Oceandy D, Morris GM, Boyett MR. Targeting miR-423-5p Reverses Exercise Training-Induced HCN4 Channel Remodeling and Sinus Bradycardia. Circ Res 2017; 121:1058-1068. [PMID: 28821541 PMCID: PMC5636198 DOI: 10.1161/circresaha.117.311607] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Revised: 08/15/2017] [Accepted: 08/17/2017] [Indexed: 11/30/2022]
Abstract
Supplemental Digital Content is available in the text. Rationale: Downregulation of the pacemaking ion channel, HCN4 (hyperpolarization-activated cyclic nucleotide gated channel 4), and the corresponding ionic current, If, underlies exercise training–induced sinus bradycardia in rodents. If this occurs in humans, it could explain the increased incidence of bradyarrhythmias in veteran athletes, and it will be important to understand the underlying processes. Objective: To test the role of HCN4 in the training-induced bradycardia in human athletes and investigate the role of microRNAs (miRs) in the repression of HCN4. Methods and Results: As in rodents, the intrinsic heart rate was significantly lower in human athletes than in nonathletes, and in all subjects, the rate-lowering effect of the HCN selective blocker, ivabradine, was significantly correlated with the intrinsic heart rate, consistent with HCN repression in athletes. Next-generation sequencing and quantitative real-time reverse transcription polymerase chain reaction showed remodeling of miRs in the sinus node of swim-trained mice. Computational predictions highlighted a prominent role for miR-423-5p. Interaction between miR-423-5p and HCN4 was confirmed by a dose-dependent reduction in HCN4 3′-untranslated region luciferase reporter activity on cotransfection with precursor miR-423-5p (abolished by mutation of predicted recognition elements). Knockdown of miR-423-5p with anti-miR-423-5p reversed training-induced bradycardia via rescue of HCN4 and If. Further experiments showed that in the sinus node of swim-trained mice, upregulation of miR-423-5p (intronic miR) and its host gene, NSRP1, is driven by an upregulation of the transcription factor Nkx2.5. Conclusions: HCN remodeling likely occurs in human athletes, as well as in rodent models. miR-423-5p contributes to training-induced bradycardia by targeting HCN4. This work presents the first evidence of miR control of HCN4 and heart rate. miR-423-5p could be a therapeutic target for pathological sinus node dysfunction in veteran athletes.
Collapse
Affiliation(s)
- Alicia D'Souza
- From the Division of Cardiovascular Sciences, University of Manchester, United Kingdom (A.D., C.M.P., Y.W., S.N., S.J.R.J.L., C.C., H.B., Y.Z., J.E., A.R., A.K., E.J.C., O.M., H.D., D.O., G.M.M., M.R.B.); K.G. Jebsen Center for Exercise in Medicine, Department of Circulation and Medical Imaging, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway (A.B.J., U.W.); Faculty of Health Sciences, NNF Center for Protein Research, University of Copenhagen, Denmark (N.L., P.C.P., A.L.); School of Healthcare Science, Manchester Metropolitan University, United Kingdom (J.C., J.M.); Department of Cardiology, CARIM School for Cardiovascular Diseases, Faculty of Health, Medicine and Life Sciences, Maastricht University, Netherlands (P.A.d.C.M.); and School of Human Movement & Nutrition Sciences, University of Queensland, Australia (U.W.)
| | - Charles M Pearman
- From the Division of Cardiovascular Sciences, University of Manchester, United Kingdom (A.D., C.M.P., Y.W., S.N., S.J.R.J.L., C.C., H.B., Y.Z., J.E., A.R., A.K., E.J.C., O.M., H.D., D.O., G.M.M., M.R.B.); K.G. Jebsen Center for Exercise in Medicine, Department of Circulation and Medical Imaging, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway (A.B.J., U.W.); Faculty of Health Sciences, NNF Center for Protein Research, University of Copenhagen, Denmark (N.L., P.C.P., A.L.); School of Healthcare Science, Manchester Metropolitan University, United Kingdom (J.C., J.M.); Department of Cardiology, CARIM School for Cardiovascular Diseases, Faculty of Health, Medicine and Life Sciences, Maastricht University, Netherlands (P.A.d.C.M.); and School of Human Movement & Nutrition Sciences, University of Queensland, Australia (U.W.)
| | - Yanwen Wang
- From the Division of Cardiovascular Sciences, University of Manchester, United Kingdom (A.D., C.M.P., Y.W., S.N., S.J.R.J.L., C.C., H.B., Y.Z., J.E., A.R., A.K., E.J.C., O.M., H.D., D.O., G.M.M., M.R.B.); K.G. Jebsen Center for Exercise in Medicine, Department of Circulation and Medical Imaging, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway (A.B.J., U.W.); Faculty of Health Sciences, NNF Center for Protein Research, University of Copenhagen, Denmark (N.L., P.C.P., A.L.); School of Healthcare Science, Manchester Metropolitan University, United Kingdom (J.C., J.M.); Department of Cardiology, CARIM School for Cardiovascular Diseases, Faculty of Health, Medicine and Life Sciences, Maastricht University, Netherlands (P.A.d.C.M.); and School of Human Movement & Nutrition Sciences, University of Queensland, Australia (U.W.)
| | - Shu Nakao
- From the Division of Cardiovascular Sciences, University of Manchester, United Kingdom (A.D., C.M.P., Y.W., S.N., S.J.R.J.L., C.C., H.B., Y.Z., J.E., A.R., A.K., E.J.C., O.M., H.D., D.O., G.M.M., M.R.B.); K.G. Jebsen Center for Exercise in Medicine, Department of Circulation and Medical Imaging, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway (A.B.J., U.W.); Faculty of Health Sciences, NNF Center for Protein Research, University of Copenhagen, Denmark (N.L., P.C.P., A.L.); School of Healthcare Science, Manchester Metropolitan University, United Kingdom (J.C., J.M.); Department of Cardiology, CARIM School for Cardiovascular Diseases, Faculty of Health, Medicine and Life Sciences, Maastricht University, Netherlands (P.A.d.C.M.); and School of Human Movement & Nutrition Sciences, University of Queensland, Australia (U.W.)
| | - Sunil Jit R J Logantha
- From the Division of Cardiovascular Sciences, University of Manchester, United Kingdom (A.D., C.M.P., Y.W., S.N., S.J.R.J.L., C.C., H.B., Y.Z., J.E., A.R., A.K., E.J.C., O.M., H.D., D.O., G.M.M., M.R.B.); K.G. Jebsen Center for Exercise in Medicine, Department of Circulation and Medical Imaging, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway (A.B.J., U.W.); Faculty of Health Sciences, NNF Center for Protein Research, University of Copenhagen, Denmark (N.L., P.C.P., A.L.); School of Healthcare Science, Manchester Metropolitan University, United Kingdom (J.C., J.M.); Department of Cardiology, CARIM School for Cardiovascular Diseases, Faculty of Health, Medicine and Life Sciences, Maastricht University, Netherlands (P.A.d.C.M.); and School of Human Movement & Nutrition Sciences, University of Queensland, Australia (U.W.)
| | - Charlotte Cox
- From the Division of Cardiovascular Sciences, University of Manchester, United Kingdom (A.D., C.M.P., Y.W., S.N., S.J.R.J.L., C.C., H.B., Y.Z., J.E., A.R., A.K., E.J.C., O.M., H.D., D.O., G.M.M., M.R.B.); K.G. Jebsen Center for Exercise in Medicine, Department of Circulation and Medical Imaging, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway (A.B.J., U.W.); Faculty of Health Sciences, NNF Center for Protein Research, University of Copenhagen, Denmark (N.L., P.C.P., A.L.); School of Healthcare Science, Manchester Metropolitan University, United Kingdom (J.C., J.M.); Department of Cardiology, CARIM School for Cardiovascular Diseases, Faculty of Health, Medicine and Life Sciences, Maastricht University, Netherlands (P.A.d.C.M.); and School of Human Movement & Nutrition Sciences, University of Queensland, Australia (U.W.)
| | - Hayley Bennett
- From the Division of Cardiovascular Sciences, University of Manchester, United Kingdom (A.D., C.M.P., Y.W., S.N., S.J.R.J.L., C.C., H.B., Y.Z., J.E., A.R., A.K., E.J.C., O.M., H.D., D.O., G.M.M., M.R.B.); K.G. Jebsen Center for Exercise in Medicine, Department of Circulation and Medical Imaging, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway (A.B.J., U.W.); Faculty of Health Sciences, NNF Center for Protein Research, University of Copenhagen, Denmark (N.L., P.C.P., A.L.); School of Healthcare Science, Manchester Metropolitan University, United Kingdom (J.C., J.M.); Department of Cardiology, CARIM School for Cardiovascular Diseases, Faculty of Health, Medicine and Life Sciences, Maastricht University, Netherlands (P.A.d.C.M.); and School of Human Movement & Nutrition Sciences, University of Queensland, Australia (U.W.)
| | - Yu Zhang
- From the Division of Cardiovascular Sciences, University of Manchester, United Kingdom (A.D., C.M.P., Y.W., S.N., S.J.R.J.L., C.C., H.B., Y.Z., J.E., A.R., A.K., E.J.C., O.M., H.D., D.O., G.M.M., M.R.B.); K.G. Jebsen Center for Exercise in Medicine, Department of Circulation and Medical Imaging, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway (A.B.J., U.W.); Faculty of Health Sciences, NNF Center for Protein Research, University of Copenhagen, Denmark (N.L., P.C.P., A.L.); School of Healthcare Science, Manchester Metropolitan University, United Kingdom (J.C., J.M.); Department of Cardiology, CARIM School for Cardiovascular Diseases, Faculty of Health, Medicine and Life Sciences, Maastricht University, Netherlands (P.A.d.C.M.); and School of Human Movement & Nutrition Sciences, University of Queensland, Australia (U.W.)
| | - Anne Berit Johnsen
- From the Division of Cardiovascular Sciences, University of Manchester, United Kingdom (A.D., C.M.P., Y.W., S.N., S.J.R.J.L., C.C., H.B., Y.Z., J.E., A.R., A.K., E.J.C., O.M., H.D., D.O., G.M.M., M.R.B.); K.G. Jebsen Center for Exercise in Medicine, Department of Circulation and Medical Imaging, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway (A.B.J., U.W.); Faculty of Health Sciences, NNF Center for Protein Research, University of Copenhagen, Denmark (N.L., P.C.P., A.L.); School of Healthcare Science, Manchester Metropolitan University, United Kingdom (J.C., J.M.); Department of Cardiology, CARIM School for Cardiovascular Diseases, Faculty of Health, Medicine and Life Sciences, Maastricht University, Netherlands (P.A.d.C.M.); and School of Human Movement & Nutrition Sciences, University of Queensland, Australia (U.W.)
| | - Nora Linscheid
- From the Division of Cardiovascular Sciences, University of Manchester, United Kingdom (A.D., C.M.P., Y.W., S.N., S.J.R.J.L., C.C., H.B., Y.Z., J.E., A.R., A.K., E.J.C., O.M., H.D., D.O., G.M.M., M.R.B.); K.G. Jebsen Center for Exercise in Medicine, Department of Circulation and Medical Imaging, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway (A.B.J., U.W.); Faculty of Health Sciences, NNF Center for Protein Research, University of Copenhagen, Denmark (N.L., P.C.P., A.L.); School of Healthcare Science, Manchester Metropolitan University, United Kingdom (J.C., J.M.); Department of Cardiology, CARIM School for Cardiovascular Diseases, Faculty of Health, Medicine and Life Sciences, Maastricht University, Netherlands (P.A.d.C.M.); and School of Human Movement & Nutrition Sciences, University of Queensland, Australia (U.W.)
| | - Pi Camilla Poulsen
- From the Division of Cardiovascular Sciences, University of Manchester, United Kingdom (A.D., C.M.P., Y.W., S.N., S.J.R.J.L., C.C., H.B., Y.Z., J.E., A.R., A.K., E.J.C., O.M., H.D., D.O., G.M.M., M.R.B.); K.G. Jebsen Center for Exercise in Medicine, Department of Circulation and Medical Imaging, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway (A.B.J., U.W.); Faculty of Health Sciences, NNF Center for Protein Research, University of Copenhagen, Denmark (N.L., P.C.P., A.L.); School of Healthcare Science, Manchester Metropolitan University, United Kingdom (J.C., J.M.); Department of Cardiology, CARIM School for Cardiovascular Diseases, Faculty of Health, Medicine and Life Sciences, Maastricht University, Netherlands (P.A.d.C.M.); and School of Human Movement & Nutrition Sciences, University of Queensland, Australia (U.W.)
| | - Jonathan Elliott
- From the Division of Cardiovascular Sciences, University of Manchester, United Kingdom (A.D., C.M.P., Y.W., S.N., S.J.R.J.L., C.C., H.B., Y.Z., J.E., A.R., A.K., E.J.C., O.M., H.D., D.O., G.M.M., M.R.B.); K.G. Jebsen Center for Exercise in Medicine, Department of Circulation and Medical Imaging, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway (A.B.J., U.W.); Faculty of Health Sciences, NNF Center for Protein Research, University of Copenhagen, Denmark (N.L., P.C.P., A.L.); School of Healthcare Science, Manchester Metropolitan University, United Kingdom (J.C., J.M.); Department of Cardiology, CARIM School for Cardiovascular Diseases, Faculty of Health, Medicine and Life Sciences, Maastricht University, Netherlands (P.A.d.C.M.); and School of Human Movement & Nutrition Sciences, University of Queensland, Australia (U.W.)
| | - Jessica Coulson
- From the Division of Cardiovascular Sciences, University of Manchester, United Kingdom (A.D., C.M.P., Y.W., S.N., S.J.R.J.L., C.C., H.B., Y.Z., J.E., A.R., A.K., E.J.C., O.M., H.D., D.O., G.M.M., M.R.B.); K.G. Jebsen Center for Exercise in Medicine, Department of Circulation and Medical Imaging, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway (A.B.J., U.W.); Faculty of Health Sciences, NNF Center for Protein Research, University of Copenhagen, Denmark (N.L., P.C.P., A.L.); School of Healthcare Science, Manchester Metropolitan University, United Kingdom (J.C., J.M.); Department of Cardiology, CARIM School for Cardiovascular Diseases, Faculty of Health, Medicine and Life Sciences, Maastricht University, Netherlands (P.A.d.C.M.); and School of Human Movement & Nutrition Sciences, University of Queensland, Australia (U.W.)
| | - Jamie McPhee
- From the Division of Cardiovascular Sciences, University of Manchester, United Kingdom (A.D., C.M.P., Y.W., S.N., S.J.R.J.L., C.C., H.B., Y.Z., J.E., A.R., A.K., E.J.C., O.M., H.D., D.O., G.M.M., M.R.B.); K.G. Jebsen Center for Exercise in Medicine, Department of Circulation and Medical Imaging, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway (A.B.J., U.W.); Faculty of Health Sciences, NNF Center for Protein Research, University of Copenhagen, Denmark (N.L., P.C.P., A.L.); School of Healthcare Science, Manchester Metropolitan University, United Kingdom (J.C., J.M.); Department of Cardiology, CARIM School for Cardiovascular Diseases, Faculty of Health, Medicine and Life Sciences, Maastricht University, Netherlands (P.A.d.C.M.); and School of Human Movement & Nutrition Sciences, University of Queensland, Australia (U.W.)
| | - Abigail Robertson
- From the Division of Cardiovascular Sciences, University of Manchester, United Kingdom (A.D., C.M.P., Y.W., S.N., S.J.R.J.L., C.C., H.B., Y.Z., J.E., A.R., A.K., E.J.C., O.M., H.D., D.O., G.M.M., M.R.B.); K.G. Jebsen Center for Exercise in Medicine, Department of Circulation and Medical Imaging, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway (A.B.J., U.W.); Faculty of Health Sciences, NNF Center for Protein Research, University of Copenhagen, Denmark (N.L., P.C.P., A.L.); School of Healthcare Science, Manchester Metropolitan University, United Kingdom (J.C., J.M.); Department of Cardiology, CARIM School for Cardiovascular Diseases, Faculty of Health, Medicine and Life Sciences, Maastricht University, Netherlands (P.A.d.C.M.); and School of Human Movement & Nutrition Sciences, University of Queensland, Australia (U.W.)
| | - Paula A da Costa Martins
- From the Division of Cardiovascular Sciences, University of Manchester, United Kingdom (A.D., C.M.P., Y.W., S.N., S.J.R.J.L., C.C., H.B., Y.Z., J.E., A.R., A.K., E.J.C., O.M., H.D., D.O., G.M.M., M.R.B.); K.G. Jebsen Center for Exercise in Medicine, Department of Circulation and Medical Imaging, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway (A.B.J., U.W.); Faculty of Health Sciences, NNF Center for Protein Research, University of Copenhagen, Denmark (N.L., P.C.P., A.L.); School of Healthcare Science, Manchester Metropolitan University, United Kingdom (J.C., J.M.); Department of Cardiology, CARIM School for Cardiovascular Diseases, Faculty of Health, Medicine and Life Sciences, Maastricht University, Netherlands (P.A.d.C.M.); and School of Human Movement & Nutrition Sciences, University of Queensland, Australia (U.W.)
| | - Ashraf Kitmitto
- From the Division of Cardiovascular Sciences, University of Manchester, United Kingdom (A.D., C.M.P., Y.W., S.N., S.J.R.J.L., C.C., H.B., Y.Z., J.E., A.R., A.K., E.J.C., O.M., H.D., D.O., G.M.M., M.R.B.); K.G. Jebsen Center for Exercise in Medicine, Department of Circulation and Medical Imaging, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway (A.B.J., U.W.); Faculty of Health Sciences, NNF Center for Protein Research, University of Copenhagen, Denmark (N.L., P.C.P., A.L.); School of Healthcare Science, Manchester Metropolitan University, United Kingdom (J.C., J.M.); Department of Cardiology, CARIM School for Cardiovascular Diseases, Faculty of Health, Medicine and Life Sciences, Maastricht University, Netherlands (P.A.d.C.M.); and School of Human Movement & Nutrition Sciences, University of Queensland, Australia (U.W.)
| | - Ulrik Wisløff
- From the Division of Cardiovascular Sciences, University of Manchester, United Kingdom (A.D., C.M.P., Y.W., S.N., S.J.R.J.L., C.C., H.B., Y.Z., J.E., A.R., A.K., E.J.C., O.M., H.D., D.O., G.M.M., M.R.B.); K.G. Jebsen Center for Exercise in Medicine, Department of Circulation and Medical Imaging, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway (A.B.J., U.W.); Faculty of Health Sciences, NNF Center for Protein Research, University of Copenhagen, Denmark (N.L., P.C.P., A.L.); School of Healthcare Science, Manchester Metropolitan University, United Kingdom (J.C., J.M.); Department of Cardiology, CARIM School for Cardiovascular Diseases, Faculty of Health, Medicine and Life Sciences, Maastricht University, Netherlands (P.A.d.C.M.); and School of Human Movement & Nutrition Sciences, University of Queensland, Australia (U.W.)
| | - Elizabeth J Cartwright
- From the Division of Cardiovascular Sciences, University of Manchester, United Kingdom (A.D., C.M.P., Y.W., S.N., S.J.R.J.L., C.C., H.B., Y.Z., J.E., A.R., A.K., E.J.C., O.M., H.D., D.O., G.M.M., M.R.B.); K.G. Jebsen Center for Exercise in Medicine, Department of Circulation and Medical Imaging, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway (A.B.J., U.W.); Faculty of Health Sciences, NNF Center for Protein Research, University of Copenhagen, Denmark (N.L., P.C.P., A.L.); School of Healthcare Science, Manchester Metropolitan University, United Kingdom (J.C., J.M.); Department of Cardiology, CARIM School for Cardiovascular Diseases, Faculty of Health, Medicine and Life Sciences, Maastricht University, Netherlands (P.A.d.C.M.); and School of Human Movement & Nutrition Sciences, University of Queensland, Australia (U.W.)
| | - Oliver Monfredi
- From the Division of Cardiovascular Sciences, University of Manchester, United Kingdom (A.D., C.M.P., Y.W., S.N., S.J.R.J.L., C.C., H.B., Y.Z., J.E., A.R., A.K., E.J.C., O.M., H.D., D.O., G.M.M., M.R.B.); K.G. Jebsen Center for Exercise in Medicine, Department of Circulation and Medical Imaging, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway (A.B.J., U.W.); Faculty of Health Sciences, NNF Center for Protein Research, University of Copenhagen, Denmark (N.L., P.C.P., A.L.); School of Healthcare Science, Manchester Metropolitan University, United Kingdom (J.C., J.M.); Department of Cardiology, CARIM School for Cardiovascular Diseases, Faculty of Health, Medicine and Life Sciences, Maastricht University, Netherlands (P.A.d.C.M.); and School of Human Movement & Nutrition Sciences, University of Queensland, Australia (U.W.)
| | - Alicia Lundby
- From the Division of Cardiovascular Sciences, University of Manchester, United Kingdom (A.D., C.M.P., Y.W., S.N., S.J.R.J.L., C.C., H.B., Y.Z., J.E., A.R., A.K., E.J.C., O.M., H.D., D.O., G.M.M., M.R.B.); K.G. Jebsen Center for Exercise in Medicine, Department of Circulation and Medical Imaging, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway (A.B.J., U.W.); Faculty of Health Sciences, NNF Center for Protein Research, University of Copenhagen, Denmark (N.L., P.C.P., A.L.); School of Healthcare Science, Manchester Metropolitan University, United Kingdom (J.C., J.M.); Department of Cardiology, CARIM School for Cardiovascular Diseases, Faculty of Health, Medicine and Life Sciences, Maastricht University, Netherlands (P.A.d.C.M.); and School of Human Movement & Nutrition Sciences, University of Queensland, Australia (U.W.)
| | - Halina Dobrzynski
- From the Division of Cardiovascular Sciences, University of Manchester, United Kingdom (A.D., C.M.P., Y.W., S.N., S.J.R.J.L., C.C., H.B., Y.Z., J.E., A.R., A.K., E.J.C., O.M., H.D., D.O., G.M.M., M.R.B.); K.G. Jebsen Center for Exercise in Medicine, Department of Circulation and Medical Imaging, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway (A.B.J., U.W.); Faculty of Health Sciences, NNF Center for Protein Research, University of Copenhagen, Denmark (N.L., P.C.P., A.L.); School of Healthcare Science, Manchester Metropolitan University, United Kingdom (J.C., J.M.); Department of Cardiology, CARIM School for Cardiovascular Diseases, Faculty of Health, Medicine and Life Sciences, Maastricht University, Netherlands (P.A.d.C.M.); and School of Human Movement & Nutrition Sciences, University of Queensland, Australia (U.W.)
| | - Delvac Oceandy
- From the Division of Cardiovascular Sciences, University of Manchester, United Kingdom (A.D., C.M.P., Y.W., S.N., S.J.R.J.L., C.C., H.B., Y.Z., J.E., A.R., A.K., E.J.C., O.M., H.D., D.O., G.M.M., M.R.B.); K.G. Jebsen Center for Exercise in Medicine, Department of Circulation and Medical Imaging, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway (A.B.J., U.W.); Faculty of Health Sciences, NNF Center for Protein Research, University of Copenhagen, Denmark (N.L., P.C.P., A.L.); School of Healthcare Science, Manchester Metropolitan University, United Kingdom (J.C., J.M.); Department of Cardiology, CARIM School for Cardiovascular Diseases, Faculty of Health, Medicine and Life Sciences, Maastricht University, Netherlands (P.A.d.C.M.); and School of Human Movement & Nutrition Sciences, University of Queensland, Australia (U.W.)
| | - Gwilym M Morris
- From the Division of Cardiovascular Sciences, University of Manchester, United Kingdom (A.D., C.M.P., Y.W., S.N., S.J.R.J.L., C.C., H.B., Y.Z., J.E., A.R., A.K., E.J.C., O.M., H.D., D.O., G.M.M., M.R.B.); K.G. Jebsen Center for Exercise in Medicine, Department of Circulation and Medical Imaging, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway (A.B.J., U.W.); Faculty of Health Sciences, NNF Center for Protein Research, University of Copenhagen, Denmark (N.L., P.C.P., A.L.); School of Healthcare Science, Manchester Metropolitan University, United Kingdom (J.C., J.M.); Department of Cardiology, CARIM School for Cardiovascular Diseases, Faculty of Health, Medicine and Life Sciences, Maastricht University, Netherlands (P.A.d.C.M.); and School of Human Movement & Nutrition Sciences, University of Queensland, Australia (U.W.)
| | - Mark R Boyett
- From the Division of Cardiovascular Sciences, University of Manchester, United Kingdom (A.D., C.M.P., Y.W., S.N., S.J.R.J.L., C.C., H.B., Y.Z., J.E., A.R., A.K., E.J.C., O.M., H.D., D.O., G.M.M., M.R.B.); K.G. Jebsen Center for Exercise in Medicine, Department of Circulation and Medical Imaging, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway (A.B.J., U.W.); Faculty of Health Sciences, NNF Center for Protein Research, University of Copenhagen, Denmark (N.L., P.C.P., A.L.); School of Healthcare Science, Manchester Metropolitan University, United Kingdom (J.C., J.M.); Department of Cardiology, CARIM School for Cardiovascular Diseases, Faculty of Health, Medicine and Life Sciences, Maastricht University, Netherlands (P.A.d.C.M.); and School of Human Movement & Nutrition Sciences, University of Queensland, Australia (U.W.).
| |
Collapse
|
47
|
Li H, Liu P, Xu S, Li Y, Dekker JD, Li B, Fan Y, Zhang Z, Hong Y, Yang G, Tang T, Ren Y, Tucker HO, Yao Z, Guo X. FOXP1 controls mesenchymal stem cell commitment and senescence during skeletal aging. J Clin Invest 2017; 127:1241-1253. [PMID: 28240601 DOI: 10.1172/jci89511] [Citation(s) in RCA: 119] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Accepted: 01/05/2017] [Indexed: 12/15/2022] Open
Abstract
A hallmark of aged mesenchymal stem/progenitor cells (MSCs) in bone marrow is the pivot of differentiation potency from osteoblast to adipocyte coupled with a decrease in self-renewal capacity. However, how these cellular events are orchestrated in the aging progress is not fully understood. In this study, we have used molecular and genetic approaches to investigate the role of forkhead box P1 (FOXP1) in transcriptional control of MSC senescence. In bone marrow MSCs, FOXP1 expression levels declined with age in an inverse manner with those of the senescence marker p16INK4A. Conditional depletion of Foxp1 in bone marrow MSCs led to premature aging characteristics, including increased bone marrow adiposity, decreased bone mass, and impaired MSC self-renewal capacity in mice. At the molecular level, FOXP1 regulated cell-fate choice of MSCs through interactions with the CEBPβ/δ complex and recombination signal binding protein for immunoglobulin κ J region (RBPjκ), key modulators of adipogenesis and osteogenesis, respectively. Loss of p16INK4A in Foxp1-deficient MSCs partially rescued the defects in replication capacity and bone mass accrual. Promoter occupancy analyses revealed that FOXP1 directly represses transcription of p16INK4A. These results indicate that FOXP1 attenuates MSC senescence by orchestrating their cell-fate switch while maintaining their replicative capacity in a dose- and age-dependent manner.
Collapse
|
48
|
Oskay Halacli S. FOXP1 enhances tumor cell migration by repression of NFAT1 transcriptional activity in MDA-MB-231 cells. Cell Biol Int 2016; 41:102-110. [PMID: 27859969 DOI: 10.1002/cbin.10702] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Accepted: 11/06/2016] [Indexed: 01/01/2023]
Abstract
Until now, forkhead box P1 (FOXP1) has been identified as a tumor suppressor in several correlation studies in breast cancer. Although FOXP1 is defined as a transcriptional repressor that interacts with other transcription factors in various mechanistic studies, there is no study that explains its repressor functions in breast cancer biology. This study demonstrated the repressor function of FOXP1 on nuclear factor of activated T cells (NFAT1) and the migratory effect of this repression in MDA-MB-231 breast cancer cells. Co-immunoprecipitation experiments were performed for the investigation of protein-protein interaction between two transcription factors. Protein-protein interaction on DNA was investigated with EMSA and transcriptional effects of FOXP1 on NFAT1, luciferase reporter assay was performed. Wound healing assay was used to analyze the effects of overexpression of FOXP1 on tumor cell migration. This study showed that FOXP1 has protein-protein interaction with NFAT1 on DNA and enhances breast cancer cell migration by repressing NFAT1 transcriptional activity and FOXP1 shows oncogenic function by regulating breast cancer cell motility.
Collapse
Affiliation(s)
- Sevil Oskay Halacli
- Department of Pediatric Immunology, Institute of Child Health, Hacettepe University, Sihhiye, 06100, Ankara, Turkey
| |
Collapse
|
49
|
Lauriol J, Cabrera JR, Roy A, Keith K, Hough SM, Damilano F, Wang B, Segarra GC, Flessa ME, Miller LE, Das S, Bronson R, Lee KH, Kontaridis MI. Developmental SHP2 dysfunction underlies cardiac hypertrophy in Noonan syndrome with multiple lentigines. J Clin Invest 2016; 126:2989-3005. [PMID: 27348588 PMCID: PMC4966304 DOI: 10.1172/jci80396] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Accepted: 05/09/2016] [Indexed: 11/17/2022] Open
Abstract
Hypertrophic cardiomyopathy is a common cause of mortality in congenital heart disease (CHD). Many gene abnormalities are associated with cardiac hypertrophy, but their function in cardiac development is not well understood. Loss-of-function mutations in PTPN11, which encodes the protein tyrosine phosphatase (PTP) SHP2, are implicated in CHD and cause Noonan syndrome with multiple lentigines (NSML), a condition that often presents with cardiac hypertrophic defects. Here, we found that NSML-associated hypertrophy stems from aberrant signaling mechanisms originating in developing endocardium. Trabeculation and valvular hyperplasia were diminished in hearts of embryonic mice expressing a human NSML-associated variant of SHP2, and these defects were recapitulated in mice expressing NSML-associated SHP2 specifically in endothelial, but not myocardial or neural crest, cells. In contrast, mice with myocardial- but not endothelial-specific NSML SHP2 expression developed ventricular septal defects, suggesting that NSML-associated mutations have both cell-autonomous and nonautonomous functions in cardiac development. However, only endothelial-specific expression of NSML-associated SHP2 induced adult-onset cardiac hypertrophy. Further, embryos expressing the NSML-associated SHP2 mutation exhibited aberrant AKT activity and decreased downstream forkhead box P1 (FOXP1)/FGF and NOTCH1/EPHB2 signaling, indicating that SHP2 is required for regulating reciprocal crosstalk between developing endocardium and myocardium. Together, our data provide functional and disease-based evidence that aberrant SHP2 signaling during cardiac development leads to CHD and adult-onset heart hypertrophy.
Collapse
Affiliation(s)
- Jessica Lauriol
- Department of Medicine, Division of Cardiology, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - Janel R. Cabrera
- Department of Medicine, Division of Cardiology, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - Ashbeel Roy
- Department of Medicine, Division of Cardiology, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - Kimberly Keith
- Department of Medicine, Division of Cardiology, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - Sara M. Hough
- Department of Medicine, Division of Cardiology, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - Federico Damilano
- Department of Medicine, Division of Cardiology, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - Bonnie Wang
- Department of Medicine, Division of Cardiology, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - Gabriel C. Segarra
- Department of Pediatrics and Department of Obstetrics and Gynecology, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Meaghan E. Flessa
- Department of Pediatrics and Department of Obstetrics and Gynecology, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Lauren E. Miller
- Department of Pediatrics and Department of Obstetrics and Gynecology, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Saumya Das
- Department of Medicine, Division of Cardiology, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | | | - Kyu-Ho Lee
- Department of Pediatrics and Department of Obstetrics and Gynecology, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Maria I. Kontaridis
- Department of Medicine, Division of Cardiology, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
50
|
|